Histone deacetylases (HDAC) regulate diverse pathways in cancer cells. Previously, we identified that Ewing sarcoma tumors, which are caused by a translocation between the EWSR1 and FLI1 genes (EWS::FLI1), are sensitive to drugs that target DNA replication, including the RRM1 and RRM2 subunits of ribonucleotide reductase, and the ATR–checkpoint kinase 1 (CHK1)–WEE1 signaling pathway. In this study, we identified that multiple HDAC inhibitors, including fimepinostat, romidepsin, and panobinostat, downregulate the levels of the RRM1, RRM2, CHK1, and WEE1 proteins in Ewing sarcoma cells and impair DNA replication. Moreover, transcriptome analyses identified that HDAC inhibitors downregulate the expression of multiple components of the prereplication complex, including the minichromosome maintenance complex 2–7 (MCM2–7) proteins and CDT1, that are essential for genomic DNA replication. Additionally, proteomic studies identified that HDAC inhibitors also downregulate the level of the BRD4 protein, a BET bromodomain protein that regulates both the transcriptional program of the EWS::FLI1 oncoprotein and DNA replication. Overall, these results provide novel insight into the molecular mechanisms by which HDAC inhibitors target cancer cells, regulate DNA replication, and inhibit the cellular response to DNA replication stress.

Significance:

In this work, we identify that HDAC inhibitors broadly disrupt DNA replication and the response to replication stress in Ewing sarcoma cells by downregulating RRM1, RRM2, CHK1, and WEE1. Notably, HDAC inhibitors also reduce MCM2–7 proteins, which are essential for prereplication complex helicase function in replication initiation and elongation, and CDT1, which loads MCM2–7 onto DNA.

The standard treatment for Ewing sarcoma—cytotoxic chemotherapy combined with surgery and/or radiation—has remained largely unchanged over the past three decades, yielding suboptimal outcomes and significant treatment-associated morbidities, such as renal toxicity, heart failure, and secondary malignancies (1, 2). This highlights the urgent need to identify tumor-specific vulnerabilities in Ewing sarcoma that can improve therapeutic outcomes and minimize toxicity.

DNA replication stress is a hallmark of cancer cells and a potential therapeutic target (37). In Ewing sarcoma tumors, DNA replication stress is driven by various factors, including cell-cycle dysregulation, increased frequency of R-loops, elevated levels of the helicase SLFN11, loss of the cohesin subunit STAG2, upregulation of nucleoside transporters, haploinsufficiency of the EWSR1 gene, functional loss of ATM activity, and a BRCA1-deficient phenotype (refs. 816; bioRxiv 2024:2023.04.30.538578). Furthermore, Ewing sarcoma cells are particularly sensitive to further exacerbation of DNA replication stress through inhibition of ribonucleotide reductase (RNR), which is the rate-limiting enzyme in the synthesis of dNTPs, and blockade of the replication stress response using ATR, checkpoint kinase 1 (CHK1), and WEE1 inhibitors (3, 1729). Clinical trials are currently evaluating combinations of these inhibitors with standard chemotherapy.

In addition to the ATR–CHK1–WEE1 axis, cells employ a preemptive strategy to mitigate replication stress by licensing dormant DNA replication origins during late M-phase and G1 phase. These dormant origins serve as backups to ensure DNA replication completion if replication forks stall or collapse (3032). The minichromosome maintenance complex 2–7 (MCM2–7) forms the ATPase core of the prereplication complex (pre-RC) helicase that licenses DNA replication origins. The MCM2–7 proteins are highly abundant in proliferating cells, and by the end of the G1 phase, the amount of DNA-bound MCM2–7 exceeds the number of active origins used during a normal S-phase (33, 34). Importantly, partial reductions in MCM2–7 levels sensitize cells to DNA replication stress without impairing normal replication (3539).

We previously demonstrated that histone deacetylase (HDAC) inhibitors downregulate the expression of the RRM1 subunit of RNR (40). In this study, we identify that HDAC inhibitors more broadly disrupt DNA replication and the cellular response to DNA replication stress by downregulating the expression of RRM1, RRM2, CHK1, and WEE1 in Ewing sarcoma cells. Notably, we also found that HDAC inhibitors reduce the levels of the MCM2–7 proteins and the CDT1 protein, which loads the MCM2–7 complex onto DNA. Finally, recognizing that HDAC inhibitors target multiple pathways beyond DNA replication, we demonstrate that BRD4, a transcriptional cofactor for the EWS::FLI1 oncoprotein and a regulator of DNA replication, and survivin (BIRC5), an antiapoptotic protein, are also targets of HDAC inhibitors in Ewing sarcoma cells (4145). Overall, these results provide novel insight into the mechanism of how histone acetylation regulates DNA replication in Ewing sarcoma tumors.

Cell lines and culture

The TC71 (RRID:CVCL_2213) and EW8 (RRID:CVCL_1658) cell lines were provided by Dr. Kimberly Stegmaier (Dana-Farber Cancer Institute, Boston, MA), the AGPN (RRID:CVCL_X981) cell line was obtained from the Childhood Cancer Repository (Children’s Oncology Group), and the HT1080 (RRID:CVCL_0317), HEK-293T (RRID:CVCL_1926), and U2OS (RRID:CVCL_0042) cell lines were obtained from ATCC. The RD (RRID:CVCL_1649) cell line was provided by Dr. Munir Tanas (University of Iowa, Iowa City, IA), and the ES6 (RRID:CVCL_1202) cell line was obtained from the Childhood Solid Tumor Network at St. Jude Children’s Research Hospital. The cell lines were maintained at 37°C in a 5% CO2 atmosphere and grown in DMEM supplemented with 10% FBS, 100 IU mL−1 penicillin, and 100 μg mL−1 streptomycin. DNA fingerprinting confirmation of cell lines was performed using the short tandem repeat method, and cell lines were used within 8 to 10 passages of thawing.

Chemical compounds

Panobinostat (HY-10224), romidepsin (HY-15149), and YM155 (HY-101-94) were obtained from MedChemExpress. Doxycycline (J67043-AE) and puromycin (J67236.8EQ) were obtained from Thermo Fisher Scientific.

Cell viability assay

Cell proliferation was measured using the alamarBlue (resazurin; Sigma-Aldrich, #R7017) fluorescence assay, as previously described (46, 47). Approximately 5 × 104 cells were plated per well of a 96-well plate, after which the cells were exposed to a range of drug concentrations for 72 hours. Fluorescence readings were then obtained after adding alamarBlue using a FLUOstar Omega microplate reader (BMG LABTECH). IC50 values were calculated using log-transformed and normalized data (GraphPad Prism 10.1.0; RRID:SCR_002798).

Protein isolation and immunoblotting

Protein extracts for immunoblotting were prepared by incubating cells in RIPA buffer (Boston BioProducts) that was supplemented with protease and phosphatase inhibitors (Halt Protease and Phosphatase Inhibitor Cocktail, EDTA-free; Thermo Fisher Scientific) for 20 minutes. Supernatants were collected following centrifugation, and SDS-PAGE was used to separate proteins, which were then transferred to polyvinylidene difluoride membranes (Millipore). Membranes were cut and divided, using a prestained protein ladder (Thermo Fisher Scientific) for molecular weight landmarks, prior to hybridization with antibodies. Chromatin-bound proteins were isolated using a Subcellular Protein Fractionation Kit for Cultured Cells (Thermo Fisher Scientific) according to the manufacturer’s instructions. Antibodies to the following proteins were used in the immunoblots: FLI1 (Abcam, #ab133485, 1:1,000, RRID:AB_2722650), actin (Cell Signaling Technology, #4970, 1:5,000; RRID:AB_2223172), α-tubulin (Proteintech, # 66031-1-Ig, 1:5,000, RRID:AB_11042766), histone H3 (Cell Signaling Technology, #4499, 1:1,000, RRID:AB_10544537), acetyl-histone H3-Lys 9 (Cell Signaling Technology, #9649, 1:1,000, RRID:AB_823528), acetyl-histone H3-Lys 14 (Cell Signaling Technology, #7627, 1:1,000, RRID:AB_10839410), acetyl-histone H3-Lys 18 (Cell Signaling Technology, #13998, 1:1,000, RRID:AB_2783723), acetyl-histone H3-Lys 27 (Cell Signaling Technology, #8173, 1:1,000, RRID:AB_10949503), acetyl-histone H3-Lys 56 (Cell Signaling Technology, #4243, 1:1,000, RRID:AB_10548193), c-myc (Cell Signaling Technology, #18583, 1:1,000, RRID:AB_2895543), BRD4 (Cell Signaling Technology, #13440, 1:1,000, RRID:AB_2687578), replication protein A (RPA2; Bethyl Laboratories, A300-244A, 1:2,000, RRID:AB_185548), MCM2 (Cell Signaling Technology, #3619, 1:1,000, RRID:AB_2142137), MCM3 (Proteintech, #15597-1-AP, 1:500, RRID:AB_2141973), MCM5 (Proteintech, #11703-1-AP, 1:1,000, RRID:AB_2235162), CDT1 (Cell Signaling Technology, #8064, 1:1,000, RRID:AB_10896851), RRM1 (Cell Signaling Technology, #8637, 1:1,000, RRID:AB_10896851), RRM2 (Proteintech, #11661-1-AP, 1:500, RRID:AB_2180392), CHK1 (Cell Signaling Technology, #2360, 1:1,000, RRID:AB_11217623), WEE1 (Cell Signaling Technology, #13084, 1:1,000, RRID:AB_2713924), and survivin (Cell Signaling Technology, #2808, 1:1,000, RRID:AB_2063948). Immunoblots were analyzed and quantified using Fiji (RRID:SCR_002285).

siRNA transfection

Cells (1.5–3 × 105) were plated 1 day prior to transfection in six-well plates. Cells were transfected with siRNA using Lipofectamine RNAiMax (Thermo Fisher Scientific) according to the manufacturer’s instructions. siControl was obtained from Cell Signaling Technology (#6568; refs. 48, 49). siMCM2 was obtained from IDT (hs.Ri.MCM2.13.1 and hs.Ri.MCM2.13.2). siHDAC1, siHDAC2, siHDAC3, and siHDAC8 were all obtained from Dharmacon (ON-TARGETplus).

RNA sequencing and analysis

RNeasy Plus Mini Kit (QIAGEN) was used to isolate RNA from cell lines. Samples were submitted to the Iowa Institute of Human Genetics Core Facility for sequencing. Samples were barcoded, pooled, and sequenced on an Illumina NovaSeq 6000 (Illumina) to obtain a minimum of 30 million paired-end 100 bp reads per sample. FastQC was used to assess the quality of the sequencing reads. Reads were then mapped against the human reference genome (hg38) using the STAR aligner (STAR, RRID:SCR_004463) and summarized at the exon level using featureCounts (RRID:SCR_012919). Raw counts were transformed using the cpm function in edgeR for sample clustering and principal component analysis. No outlier samples were identified or removed from the analysis. The DESeq2 package (DESeq, RRID:SCR_000154) was used for the identification of differentially expressed genes. Differentially expressed gene data were analyzed for gene set enrichment and transcription factor enrichment analysis using ShinyGO 0.77 (RRID:SCR_019213), Enrichr (RRID:SCR_001575), and iDEP.96 (5052). The sequencing data are available on the Gene Expression Omnibus under the accession number GSE280573. Previously published RNA sequencing (RNA-seq) data for Ewing sarcoma cell lines treated with siRNA targeting EWS::FLI1 are available on the Gene Expression Omnibus under the accession number GSE263504 (53). Previously published RNA-seq data for Ewing sarcoma cell lines with CRISPR-mediated knockout of RRM1 are available on the Gene Expression Omnibus under the accession number GSE215881.

qRT-PCR

Total RNA was extracted from cells using an RNeasy kit (QIAGEN) following the manufacturer’s instructions. One microgram of total RNA was reverse-transcribed into first-strand cDNA using random hexamer primers and SuperScript III Reverse Transcriptase (Thermo Fisher Scientific). qRT-PCR was performed on the ViiA 7 Real-Time PCR System (Life Technologies) using SYBR Select Master Mix (Thermo Fisher Scientific). The qRT-PCR primers for BRD4, EWS::FLI1, and GAPDH were 5′-GAG​CTA​CCC​ACA​GAA​GAA​ACC-3′, 5′-TCC​TAC​AGC​CAA​GCT​CCA​AGT​C-3′, and 5′-CTG​GGC​TAC​ACT​GAG​CAC​C-3′, respectively. Reactions were performed in triplicate, and gene expression was normalized to GAPDH.

EdU labeling and detection

Detection of DNA replication was performed in triplicate using a Click-iT 5-ethynyl-2′-deoxyuridine (EdU)-488 kit (Thermo Fisher Scientific; ref. 17). Briefly, cells were labeled with 10 mmol/L EdU for 1 hour and then harvested using trypsin and fixed using the Click-iT fixative buffer. Cells were then washed by centrifugation and resuspended in Click-iT saponin-based permeabilization and wash reagent. Click-iT Plus reaction cocktail was added to the cell suspension for 30 minutes, after which the cells were washed by centrifugation. Flow cytometry was performed on a BD LSR II flow cytometer.

Doxycycline-inducible shRPA2

shERWOOD UltramiR lentiviral-inducible short hairpin RNA (shRNA) plasmids targeting shRPA2 were obtained from transOMIC Technologies. Lentivirus was produced by transfecting HEK-293T cells with the shRNA plasmid and packaging plasmids (psPAX2 and pMD2.G) according to the FuGENE 6 (Roche) protocol. For the lentiviral transduction, Ewing sarcoma cells were incubated with 2 mL of virus and 6 mg/mL of polybrene (Sigma-Aldrich) for 12 to 16 hours. Cells were selected in 1 μg/mL puromycin 48 hours after transduction.

RPPA

Reverse phase protein array (RPPA) analysis of cell lines was performed by the RPPA Core Facility at the MD Anderson Cancer Center. Cells were provided to the core facility as frozen pellets, and the protein extraction, data normalization, and analysis were performed according to facility protocols (https://www.mdanderson.org/research/research-resources/core-facilities/functional-proteomics-rppa-core/education-and-references.html; RRID:SCR_016649).

Cancer Dependency Map

Gene expression and CRISPR dependency data from the Cancer Dependency Map resource were accessed through the DepMap Portal (depmap.org; RRID:SCR_017655; refs. 54, 55).

Survival analysis

Kaplan–Meier curves were analyzed for significance using the log-rank (Mantel–Cox) test with the R2 Genomics Platform (https://r2.amc.nl; RRID:SCR_025770) and the Ewing Sarcoma (Savola) dataset (56).

Statistical analysis

Statistical analyses were conducted using GraphPad Prism 10.1.0 (RRID: SCR_002798). Single comparisons were analyzed by the unpaired two-tailed t test. Multiple comparisons were analyzed by one-way ANOVA with the Tukey post hoc multiple comparisons test. P values of less than or equal to 0.05 were considered significant.

Data availability

Raw data are available in the Supplementary Supporting Data Values file. Unedited immunoblots are available in the unedited blot and gel images file. The sequencing data are available on the Gene Expression Omnibus under the accession number GSE280573. Previously published RNA-seq data for Ewing sarcoma cell lines treated with siRNA targeting EWS::FLI1 are available on the Gene Expression Omnibus under the accession number GSE263504 (53). Previously published RNA-seq data for Ewing sarcoma cell lines with CRISPR-mediated knockout of RRM1 are available on the Gene Expression Omnibus under the accession number GSE215881.

HDAC inhibitors decrease RRM1, RRM2, CHK1, and WEE1 protein levels

Ewing sarcoma cells are sensitive to the dual inhibition of RNR and CHK1, a key mediator of the response to DNA replication stress (17, 1922). Previously, we identified that HDAC inhibitors downregulate the RNR subunit RRM1 protein expression in Ewing sarcoma cells (40). Notably, fimepinostat, a dual inhibitor of HDAC and PI3K, was recently shown in leukemia cell lines to reduce the levels of additional proteins with critical roles in DNA replication, including RRM2, CHK1, and WEE1 (57, 58). Consequently, we began this work by testing whether fimepinostat targets DNA replication and replication stress response proteins in Ewing sarcoma cells.

Figure 1A shows that the treatment of Ewing sarcoma cell lines, EW8 and TC71, with fimepinostat for 24 hours reduces the levels of RRM1, RRM2, CHK1, and WEE1. To determine whether this effect of fimepinostat was due to inhibition of HDAC, PI3K, or both targets, we treated the cell lines with the PI3K inhibitor LY3023414. Figure 1B demonstrates that LY3023414 did not alter the levels of the replication proteins, suggesting that HDAC inhibition is the more relevant target for regulation of the DNA replication and replication stress proteins. Next, we treated the Ewing sarcoma cell lines with low doses of panobinostat (pan-HDAC inhibitor) or romidepsin (an inhibitor of class 1 HDACs: HDAC1, HDAC2, HDAC3, and HDAC8) and assessed acetylation at multiple sites in the histone H3 protein. Figure 1C shows that both drugs, at doses of 5 to 10 nmol/L, increase histone acetylation. Dose–response experiments were then performed with both drugs to evaluate the effect of HDAC inhibition on RRM1, RRM2, CHK1, and WEE1 protein levels. Figure 1D and E demonstrate that panobinostat and romidepsin decrease the levels of RRM1, RRM2, CHK1, and WEE1 proteins while increasing the phosphorylation of γgH2AX, a marker of DNA damage.

Figure 1

HDAC inhibitors decrease RRM1, RRM2, CHK1, and WEE1 protein levels. A, EW8 and TC71 cells were treated with fimepinostat (25 or 100 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting. B, EW8 and TC71 cells were treated with LY3023414 (1,000 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting. C, EW8 and TC71 cells were treated with panobinostat (10 nmol/L) or romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting. D and E, EW8 cells were treated with different doses of panobinostat (D) or romidepsin (E) for 24 hours, and then cellular lysates were collected for immunoblotting. F, Cell lines representing additional sarcoma subtypes were treated with romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting.

Figure 1

HDAC inhibitors decrease RRM1, RRM2, CHK1, and WEE1 protein levels. A, EW8 and TC71 cells were treated with fimepinostat (25 or 100 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting. B, EW8 and TC71 cells were treated with LY3023414 (1,000 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting. C, EW8 and TC71 cells were treated with panobinostat (10 nmol/L) or romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting. D and E, EW8 cells were treated with different doses of panobinostat (D) or romidepsin (E) for 24 hours, and then cellular lysates were collected for immunoblotting. F, Cell lines representing additional sarcoma subtypes were treated with romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting.

Close modal

Dose–response experiments demonstrated that HDAC inhibitors decrease Ewing sarcoma cell proliferation in 72-hour cell growth assays, consistent with the established role of these genes in cell survival and published work on the sensitivity of Ewing sarcoma cell lines to HDAC inhibitors (Supplementary Fig. S1; refs. 5964). Ewing sarcoma cell lines were also previously shown to be sensitive to the CRISPR/Cas9-mediated knockout of class 1 HDACs, including HDAC1, HDAC2, and HDAC3 (59). Analysis of the Dependency Map (Broad Institute) data identified that the mRNA expression levels of HDAC1, HDAC2, and HDAC3, compared with class 2 HDACs, strongly correlate with the expression levels of the DNA replication genes (Supplementary Fig. S2A–S2C). We used siRNA to knock down individual class 1 HDACs but found that downregulation of one HDAC resulted in upregulation of other HDACs (Supplementary Fig. S3A). This compensatory response of class 1 HDACs was also reported in CRISPR/Cas9-mediated knockout experiments (59). Consequently, to further evaluate the effect of inhibiting multiple class 1 HDACs, we tested a second class 1 HDAC inhibitor, entinostat, and observed similar effects to romidepsin (Supplementary Fig. S3B). Finally, we treated additional sarcoma cell lines, including HT1080 (fibrosarcoma), RD (rhabdomyosarcoma), and U2OS (osteosarcoma), with romidepsin and observed a similar decrease in the levels of multiple replication proteins, demonstrating that the effect of HDAC inhibitors on DNA replication proteins is not limited to Ewing sarcoma tumors (Fig. 1F).

HDAC inhibitors downregulate the expression of genes related to DNA replication and the cellular response to DNA replication stress

Next, we investigated how HDAC proteins, which are epigenetic regulators of gene expression, regulate the levels of DNA replication proteins in Ewing sarcoma cells (65, 66). EW8 cells were treated with multiple doses of panobinostat (5 and 10 nmol/L) or romidepsin (2.5 and 5 nmol/L), which reduced levels of the DNA replication proteins, and then RNA-seq analysis was performed to assess the effect of HDAC inhibition on the transcriptome (Fig. 2A). Volcano plots for the two drugs show that the treatment of Ewing sarcoma cells with HDAC inhibitors results in more upregulated genes than downregulated genes, consistent with the well-established effect of histone acetylation on chromatin structure and accessibility (Fig. 2B and C; Supplementary Fig. S4; ref. 67). However, as shown in Fig. 2D and E, both HDAC inhibitors significantly downregulated the expression of the DNA replication–related genes RRM1, RRM2, CHK1, and WEE1. Moreover, gene set enrichment analysis, using the Kyoto Encyclopedia of Genes and Genomes pathway dataset, identified DNA replication as a top gene set downregulated by both drugs (Fig. 2F and G). Further analysis of the genes comprising these enriched gene sets unexpectedly identified that romidepsin and panobinostat also downregulate multiple components of the MCM2–7, which is the ATPase core of the pre-RC that licenses DNA replication origins (Fig. 2H; Supplementary Fig. S5; ref. 34). HDAC inhibitors decreased MCM2–7 expression by ∼2- to 4-fold (Fig. 2H), and Fig. 2I demonstrates that the levels of the MCM2–7 proteins are also decreased by treatment with romidepsin. Additionally, the level of CDT1, which loads the MCM2–7 complex onto DNA, was also decreased by treatment with an HDAC inhibitor. This effect on MCM2–7 and CDT1 expression is not a general response to DNA replication stress as previous RNA-seq analysis of EW8 and TC71 cell lines with conditional knockout and rescue of RRM1, which arrests DNA replication, did not show a similar effect on MCM2–7 or CDT1 mRNA levels (Supplementary Fig. S6; ref. 40).

Figure 2

HDAC inhibitors downregulate the expression of genes related to DNA replication and the cellular response to DNA replication stress. A, EW8 cells were treated with panobinostat or romidepsin for 24 hours, and then cellular lysates and mRNA were collected for immunoblotting and RNA-seq analysis. B and C, Volcano plots of differentially expressed genes (fold change >2, adjusted P value < 0.05) in the EW8 cells treated with romidepsin (5 nmol/L; B) or panobinostat (10 nmol/L; C). D–E, RNA-seq data, fold change and adjusted P value, for the RRM1, RRM2, CHK1, and WEE1 genes in EW8 cells treated with romidepsin (5 nmol/L) and panobinostat (10 nmol/L). F and G, Gene sets (Kyoto Encyclopedia of Genes and Genomes pathway) enriched in the genes that are downregulated in EW8 cells treated with romidepsin (F) and panobinostat (G). H, RNA-seq data, fold change and adjusted P value, for the MCM2–7 genes in EW8 cells treated with romidepsin and panobinostat. I, EW8 and TC71 cells were treated with romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting.

Figure 2

HDAC inhibitors downregulate the expression of genes related to DNA replication and the cellular response to DNA replication stress. A, EW8 cells were treated with panobinostat or romidepsin for 24 hours, and then cellular lysates and mRNA were collected for immunoblotting and RNA-seq analysis. B and C, Volcano plots of differentially expressed genes (fold change >2, adjusted P value < 0.05) in the EW8 cells treated with romidepsin (5 nmol/L; B) or panobinostat (10 nmol/L; C). D–E, RNA-seq data, fold change and adjusted P value, for the RRM1, RRM2, CHK1, and WEE1 genes in EW8 cells treated with romidepsin (5 nmol/L) and panobinostat (10 nmol/L). F and G, Gene sets (Kyoto Encyclopedia of Genes and Genomes pathway) enriched in the genes that are downregulated in EW8 cells treated with romidepsin (F) and panobinostat (G). H, RNA-seq data, fold change and adjusted P value, for the MCM2–7 genes in EW8 cells treated with romidepsin and panobinostat. I, EW8 and TC71 cells were treated with romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting.

Close modal

HDAC inhibitors impair DNA replication in Ewing sarcoma cells

The downregulation of these pre-RC proteins is predicted to impair DNA replication and generate replication stress (33, 34). Figure 3A and B shows that romidepsin inhibits DNA replication, as assessed by measuring the incorporation efficiency of the thymidine analog EdU in multiple Ewing sarcoma cell lines. Moreover, DNA replication stress generates ssDNA, which is bound by RPA2, and Fig. 3C demonstrates that HDAC inhibitors increase chromatin-bound RPA2 (68). Reducing RPA2 levels using doxycycline-inducible shRNA also sensitizes Ewing sarcoma cells to romidepsin and panobinostat (Fig. 3D–F). Analysis of Dependency Map (Broad Institute) data identified that the loss of MCM2–7 proteins impairs proliferation in Ewing sarcoma and other cancer types (Fig. 3G; ref. 54). Similarly, knockdown of MCM2 using two different siRNAs inhibits DNA replication, as assessed using EdU labeling (Fig. 3H and I). Finally, high levels of CDT1 and the individual MCM2–7 proteins, which are associated with poor prognoses in multiple cancer types, are significantly associated with poor survival in patients with Ewing sarcoma tumors (Fig. 3J; Supplementary Fig. S7; refs. 69, 70).

Figure 3

HDAC inhibitors impair DNA replication in Ewing sarcoma cells. A, Representative cell-cycle analysis (EdU and propidium iodide) plot for EW8 cells treated with romidepsin (5 nmol/L) for 24 hours. B, Summary of EdU incorporation efficiency for additional Ewing sarcoma cell lines treated with romidepsin (5 nmol/L) for 24 hours. C, Ewing sarcoma cell lines were treated with romidepsin (5 nmol/L) for 24 hours, and then chromatin-bound RPA2 was assessed using immunoblotting. D, EW8 cells expressing a doxycycline-inducible shRNA targeting RPA2 were treated with doxycycline for 24 hours, and then cellular lysates were collected for immunoblotting. E and F, EW8 cells expressing a doxycycline-inducible shRNA targeting RPA2 were treated with doxycycline (1,000 ng/mL) for 24 hours, and then dose-response experiments were performed with romidepsin (E) and panobinostat (F). Cell viability was measured 72 hours after drug addition using the alamarBlue assay. Error bars represent the mean ± SD of three technical replicates. The results are representative of two independent experiments. G, CRISPR dependency score for CDT1 and MCM2–7 (depmap.org). H, EW8 and TC71 cells were treated with two different siRNAs targeting MCM2, and then cellular lysates were collected for immunoblotting. I, EW8 and TC71 cells were treated with siRNAs targeting MCM2, and then EdU incorporation into DNA was measured using flow cytometry. J, Kaplan–Meier analysis shows overall survival of patients with Ewing sarcoma tumors according to the expression level of CDT1 and MCM2 mRNA. P values were calculated using a two-tailed Student t test or a one-way ANOVA followed by the Dunnett multiple comparisons test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Figure 3

HDAC inhibitors impair DNA replication in Ewing sarcoma cells. A, Representative cell-cycle analysis (EdU and propidium iodide) plot for EW8 cells treated with romidepsin (5 nmol/L) for 24 hours. B, Summary of EdU incorporation efficiency for additional Ewing sarcoma cell lines treated with romidepsin (5 nmol/L) for 24 hours. C, Ewing sarcoma cell lines were treated with romidepsin (5 nmol/L) for 24 hours, and then chromatin-bound RPA2 was assessed using immunoblotting. D, EW8 cells expressing a doxycycline-inducible shRNA targeting RPA2 were treated with doxycycline for 24 hours, and then cellular lysates were collected for immunoblotting. E and F, EW8 cells expressing a doxycycline-inducible shRNA targeting RPA2 were treated with doxycycline (1,000 ng/mL) for 24 hours, and then dose-response experiments were performed with romidepsin (E) and panobinostat (F). Cell viability was measured 72 hours after drug addition using the alamarBlue assay. Error bars represent the mean ± SD of three technical replicates. The results are representative of two independent experiments. G, CRISPR dependency score for CDT1 and MCM2–7 (depmap.org). H, EW8 and TC71 cells were treated with two different siRNAs targeting MCM2, and then cellular lysates were collected for immunoblotting. I, EW8 and TC71 cells were treated with siRNAs targeting MCM2, and then EdU incorporation into DNA was measured using flow cytometry. J, Kaplan–Meier analysis shows overall survival of patients with Ewing sarcoma tumors according to the expression level of CDT1 and MCM2 mRNA. P values were calculated using a two-tailed Student t test or a one-way ANOVA followed by the Dunnett multiple comparisons test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Close modal

HDAC inhibitors upregulate and downregulate genes that are regulated by EWS::FLI1

Next, we analyzed the gene expression data using transcription factor enrichment analysis and identified that HDAC inhibitors upregulate and downregulate genes that are also regulated by EWS::FLI1 (Fig. 4A and B). Previously, in published work, we used siRNA to knock down EWS::FLI1 and then performed RNA-seq analysis of the transcriptome (53). Supporting the transcription factor enrichment analysis, Fig. 4C and D demonstrates that HDAC inhibitors regulate a significant number of genes that are also regulated by EWS::FLI1 in the siRNA knockdown experiment. Although HDAC inhibitors have been reported to inhibit the expression of multiple oncogenes, including EWS::FLI1, we found that under the conditions tested, using low doses of HDAC inhibitors, EWS::FLI1 mRNA was not significantly altered in either cell line, and the EWS-FLI1 protein was only modestly decreased in one cell line (Fig. 4E and F; refs. 71, 72).

Figure 4

HDAC inhibitors upregulate and downregulate genes that are regulated by EWS::FLI1. A and B, Results of transcription factor enrichment analysis performed with the genes that are upregulated (A) and downregulated (B) in EW8 cells treated with romidepsin. C and D, Overlap between genes upregulated (C) and downregulated (D) in EW8 cells treated with romidepsin or siEWS::FLI1. E, Relative EWS::FLI1 mRNA in EW8 and TC71 cells treated with romidepsin for 24 hours. F, EW8 and TC71 cells were treated with romidepsin or panobinostat for 24 hours, and then cellular lysates were collected for immunoblotting for the EWS::FLI1 oncoprotein.

Figure 4

HDAC inhibitors upregulate and downregulate genes that are regulated by EWS::FLI1. A and B, Results of transcription factor enrichment analysis performed with the genes that are upregulated (A) and downregulated (B) in EW8 cells treated with romidepsin. C and D, Overlap between genes upregulated (C) and downregulated (D) in EW8 cells treated with romidepsin or siEWS::FLI1. E, Relative EWS::FLI1 mRNA in EW8 and TC71 cells treated with romidepsin for 24 hours. F, EW8 and TC71 cells were treated with romidepsin or panobinostat for 24 hours, and then cellular lysates were collected for immunoblotting for the EWS::FLI1 oncoprotein.

Close modal

HDAC inhibitors decrease BRD4 protein levels in Ewing sarcoma cells

Next, we performed RPPA analysis of Ewing sarcoma (EW8, TC71, and ES6) and U2OS (osteosarcoma) cell lines treated with panobinostat and romidepsin (Fig. 5A and B; Supplementary Fig. S8). Notably, a single protein, BRD4, was decreased >2-fold in all datasets (Fig. 5C; Supplementary Table S1). BRD4 is a transcriptional and epigenetic regulator that recognizes acetylated lysine residues (73). In addition, BRD4 is also a well-described coregulator of the EWS::FLI1 transcriptional program (4345, 74). HDAC inhibitors did not decrease BRD4 mRNA, but immunoblotting confirmed the RPPA results, showing downregulation of BRD4 protein levels (Fig. 5D and E). BRD4 is reported to support cell proliferation in multiple cancer types, and analysis of Dependency Map (Broad Institute) data identified that reducing BRD4 levels, by shRNA knockdown or CRISPR knockout, inhibits the proliferation of Ewing sarcoma and other cancer cell lines (Fig. 5F). BRD4 is also reported to regulate DNA replication and the cellular response to DNA replication stress via multiple targets and mechanisms (7577). Figure 5G–I demonstrate that decreasing BRD4 protein levels, using dBET1 (a small-molecule BRD4 degrader), inhibits DNA replication as assessed by EdU labeling of DNA.

Figure 5

HDAC inhibitors decrease BRD4 protein levels in Ewing sarcoma cells. A and B, Comparison of protein expression, based on RPPAs, in EW8 (A) or TC71 (B) cells treated with romidepsin (5 nmol/L) or panobinostat (10 nmol/L) for 24 hours. C, Overlap of proteins downregulated >1.5-fold in the RPPA experiment by romidepsin and panobinostat in Ewing sarcoma (ES6, EW8, and TC71) and osteosarcoma (U2OS) cell lines. D, Relative BRD4 mRNA in EW8 and TC71 cells treated with romidepsin (5 nmol/L). E, EW8 and TC71 cells were treated with romidepsin or panobinostat for 24 hours, and then cellular lysates were collected for immunoblotting for BRD4. F, CRISPR and shRNA dependency scores for BRD4 (Dependency Map, Broad Institute) in Ewing sarcoma and other cancer cell lines. G, EW8 and TC71 cells were treated with dBET1 (5 μmol/L) for 18 hours, and then cellular lysates were collected for immunoblotting for BRD4. H, Representative cell-cycle analysis (EdU and propidium iodide) plot for EW8 cells treated with dBET1 (5 µmol/L) for 18 hours. I, Summary of EdU incorporation into DNA for additional Ewing sarcoma cell lines treated with dBET1 (5 µmol/L) for 18 hours. P values were calculated using a two-tailed Student t test. **, P < 0.01. FC, fold change.

Figure 5

HDAC inhibitors decrease BRD4 protein levels in Ewing sarcoma cells. A and B, Comparison of protein expression, based on RPPAs, in EW8 (A) or TC71 (B) cells treated with romidepsin (5 nmol/L) or panobinostat (10 nmol/L) for 24 hours. C, Overlap of proteins downregulated >1.5-fold in the RPPA experiment by romidepsin and panobinostat in Ewing sarcoma (ES6, EW8, and TC71) and osteosarcoma (U2OS) cell lines. D, Relative BRD4 mRNA in EW8 and TC71 cells treated with romidepsin (5 nmol/L). E, EW8 and TC71 cells were treated with romidepsin or panobinostat for 24 hours, and then cellular lysates were collected for immunoblotting for BRD4. F, CRISPR and shRNA dependency scores for BRD4 (Dependency Map, Broad Institute) in Ewing sarcoma and other cancer cell lines. G, EW8 and TC71 cells were treated with dBET1 (5 μmol/L) for 18 hours, and then cellular lysates were collected for immunoblotting for BRD4. H, Representative cell-cycle analysis (EdU and propidium iodide) plot for EW8 cells treated with dBET1 (5 µmol/L) for 18 hours. I, Summary of EdU incorporation into DNA for additional Ewing sarcoma cell lines treated with dBET1 (5 µmol/L) for 18 hours. P values were calculated using a two-tailed Student t test. **, P < 0.01. FC, fold change.

Close modal

HDAC inhibitors downregulate survivin (BIRC5) in Ewing sarcoma cells

Although our analysis focused on DNA replication, we expect that the effects of HDAC inhibitors affect additional pathways and targets beyond DNA replication. For example, gene set enrichment analysis (Hallmark dataset) of the genes downregulated by HDAC inhibitors identified enrichment for c-Myc targets. Immunoblotting validated that HDAC inhibitors downregulate the expression level of the c-Myc protein, which supports Ewing sarcoma tumorigenesis (Supplementary Fig. S9; refs. 78, 79). We also observed drug-induced changes in cell morphology suggestive of apoptosis (Fig. 6A), and Figure 6B shows that romidepsin treatment causes cleavage of PARP, a marker of apoptosis. Analysis of the RNA-seq data identified a ∼2-fold decrease in survivin (BIRC5), an antiapoptotic protein with a prosurvival function in Ewing sarcoma tumors (Fig. 6C; refs. 41, 42). Treatment of multiple Ewing sarcoma cell lines with romidepsin reduced the level of the survivin protein (Fig. 6D). Survivin is highly expressed in Ewing sarcoma cell lines, and the loss of survivin significantly decreases cell proliferation (Fig. 6E; refs. 41, 42, 80). Next, we treated EW8 cells with YM155, a drug that suppresses survivin expression (Fig. 6F), and identified a significant inhibition of cell growth (Fig. 6G). Finally, high levels of survivin are associated with poor survival in patients with Ewing sarcoma tumors (Fig. 6H; refs. 42, 80).

Figure 6

HDAC inhibitors downregulate survivin (BIRC5) in Ewing sarcoma cells. A, Treatment with romidepsin (5 nmol/L) causes morphologic changes in Ewing sarcoma cells. B, EW8 and TC71 cells were treated with romidepsin for 24 or 30 hours, and then cellular lysates were collected for immunoblotting. C, RNA-seq data, fold change and adjusted P value, for the survivin gene in EW8 cells treated with romidepsin and panobinostat. D, Ewing sarcoma cell lines were treated with romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting for survivin. E, Survivin mRNA expression level and CRISPR dependency score in Ewing sarcoma, other sarcoma, and other cancer cell lines. F, EW8 and TC71 cells were treated with YM155 for 24 hours, and then cellular lysates were collected for immunoblotting for survivin. G, EW8 cells were treated with different doses of YM115 for 72 hours, and then cell viability was measured using the alamarBlue assay. Error bars represent the mean ± SD of three technical replicates. The results are representative of two independent experiments. H, Kaplan–Meier analysis shows the overall survival of patients with Ewing sarcoma according to the expression level of survivin mRNA. P values were calculated using a one-way ANOVA followed by the Dunnett multiple comparisons test. TPM, transcripts per million. ***, P < 0.001.

Figure 6

HDAC inhibitors downregulate survivin (BIRC5) in Ewing sarcoma cells. A, Treatment with romidepsin (5 nmol/L) causes morphologic changes in Ewing sarcoma cells. B, EW8 and TC71 cells were treated with romidepsin for 24 or 30 hours, and then cellular lysates were collected for immunoblotting. C, RNA-seq data, fold change and adjusted P value, for the survivin gene in EW8 cells treated with romidepsin and panobinostat. D, Ewing sarcoma cell lines were treated with romidepsin (5 nmol/L) for 24 hours, and then cellular lysates were collected for immunoblotting for survivin. E, Survivin mRNA expression level and CRISPR dependency score in Ewing sarcoma, other sarcoma, and other cancer cell lines. F, EW8 and TC71 cells were treated with YM155 for 24 hours, and then cellular lysates were collected for immunoblotting for survivin. G, EW8 cells were treated with different doses of YM115 for 72 hours, and then cell viability was measured using the alamarBlue assay. Error bars represent the mean ± SD of three technical replicates. The results are representative of two independent experiments. H, Kaplan–Meier analysis shows the overall survival of patients with Ewing sarcoma according to the expression level of survivin mRNA. P values were calculated using a one-way ANOVA followed by the Dunnett multiple comparisons test. TPM, transcripts per million. ***, P < 0.001.

Close modal

The EWS::FLI1 gene fusion is the driver mutation in most Ewing sarcoma tumors and functions, in part, as an aberrant transcription factor (81). EWS::FLI1 is an attractive therapeutic target in Ewing sarcoma tumors because it is required for tumorigenesis, but directly targeting this oncoprotein has been challenging despite significant efforts over the past three decades (82). A complementary therapeutic approach is to identify and target unique vulnerabilities incurred by the oncoprotein. Notably, our group and others have shown that Ewing sarcoma tumors are sensitive, in vitro and in vivo in xenograft tumors, to drugs targeting the DNA replication pathway, including drug combinations targeting RNR and the ATR–CHK1 signaling pathway (refs. 3, 1729; bioRxiv 2024:2023.04.30.538578).

In this study, we identified that HDAC inhibitors decrease the mRNA and protein levels of multiple proteins, including RRM1, RRM2, CHK1, and WEE1, that have critical roles in DNA replication and the response to DNA replication stress (Fig. 7). Unexpectedly, our RNA-seq and proteomic analyses also revealed that HDAC inhibitors decrease the expression of the pre-RC components MCM2–7 and CDT1, as well as BRD4, a transcriptional cofactor for the EWS::FLI1 oncoprotein (Fig. 7).

Figure 7

Integrated model for the regulation of DNA replication and replication stress response pathways by HDAC inhibitors. HDAC inhibitors downregulate the expression of multiple components of the pre-RC, including the MCM2–7 proteins and CDT1, as well as the RRM1 and RRM2 subunits of RNR, CHK1, and WEE1. HDAC inhibitors also downregulate the levels of the BRD4 and survivin proteins.

Figure 7

Integrated model for the regulation of DNA replication and replication stress response pathways by HDAC inhibitors. HDAC inhibitors downregulate the expression of multiple components of the pre-RC, including the MCM2–7 proteins and CDT1, as well as the RRM1 and RRM2 subunits of RNR, CHK1, and WEE1. HDAC inhibitors also downregulate the levels of the BRD4 and survivin proteins.

Close modal

The strategy of targeting the pre-RC to enhance the efficacy of drugs targeting DNA replication is unexplored compared with efforts that have focused on understanding how the DNA damage response kinases, including ATR and CHK1, mediate resistance to drugs that cause or exacerbate DNA replication stress. Moreover, translational methods to target the MCM2–7 proteins in the clinic are lacking (30, 31, 70). However, in this work, we identified that HDAC inhibitors downregulate the expression of the MCM2–7 proteins, as well as the CDT1 protein that loads MCM2–7 onto the DNA. As MCM2–7 is loaded onto DNA in late mitosis and the G1 phase of the cell cycle, HDAC inhibitors may provide a novel approach to preemptively sensitize cells to DNA replication stress and, thereby, target DNA replication in cells before DNA replication begins (30, 35, 83).

Further supporting a critical role for the MCM2–7 and the pre-RC in Ewing sarcoma tumors, the Dbf4-dependent Cdc7 kinase (DDK), which initiates replisome assembly by phosphorylating the N-terminal tails of Mcm2, Mcm4, and Mcm6, was recently identified as a target in Ewing sarcoma tumors (3, 26, 27). Treatment of Ewing sarcoma cell lines with DDK inhibitors reduced the rate of DNA replication, prolonged the S-phase, induced aberrant mitotic progression, and demonstrated synergy with WEE1 inhibitors (3, 26, 27). In future work, we plan to test for synergy between HDAC and DDK inhibitors, as well as assess the effects of the drug combination on replisome function.

HDAC inhibitors are known to downregulate the expression of multiple oncogenes, including EWS::FLI1 (71, 72). The effects of HDAC inhibitors on the transcription of the EWS::FLI1 oncogene are dose-dependent with downregulation of EWS::FLI1 occurring at higher drug doses (71). In our experiments, using low doses of HDAC inhibitors (<10 nmol/L), we did not observe an effect of HDAC inhibitors on EWS::FLI1 levels. However, we expect that the modulation of EWS::FLI1 levels by HDAC inhibitors will be dependent on multiple factors, including the specific cell line, HDAC inhibitor, duration of treatment, and drug dose. Although low-dose HDAC inhibitors did not affect the level of EWS::FLI1 in our experiments, proteomic analyses identified that HDAC inhibitors do reduce the level of the BRD4 protein in Ewing sarcoma cell lines. BRD4 is a transcriptional regulator that recognizes acetylated lysine residues and functions as a coregulator for the transcriptional program of the EWS::FLI1 oncoprotein (4345, 74). Thus, the loss of BRD4 is expected to contribute to the overlap in gene expression signatures that we identified between HDAC inhibitors and the shRNA-mediated downregulation of EWS::FLI1.

HDAC inhibition is also reported to increase histone 4 polyacetylation, which is a preferred binding substrate of BRD4, and retarget BRD4 to the bodies of actively transcribed genes (84). This redistribution of BRD4 could further disrupt the function of BRD4 in Ewing sarcoma cells and exacerbate the effects of HDAC inhibitors on the decrease of BRD4 protein levels. Moreover, BRD4 has also been reported to regulate multiple steps of DNA replication and the response to DNA replication stress, including activation of the ATR–CHK1 pathway and resolution of R-loops. R-loops, which are upregulated in Ewing sarcoma tumors, are DNA–RNA hybrids that impair DNA replication and cause DNA replication stress (7577). We hypothesize that the loss of BRD4 mediated by HDAC inhibitors could further upregulate R-loops in Ewing sarcoma tumors, which will be tested in future work. In addition, recent work has also identified that inhibition of HDAC8 activity can upregulate R-loops in cancer cells (85).

HDAC inhibitors exhibit pleiotropic effects in cancer cells, targeting diverse pathways that are critical for tumorigenesis (59, 6264). Although our study focused on the effects of HDAC inhibitors on DNA replication, we also identified additional targets and pathways, including survivin and c-Myc, that support tumorigenesis in Ewing sarcoma tumors and are regulated by HDAC inhibitors. Elucidating these additional pathways will be crucial for identifying drugs that synergize with HDAC inhibitors and advancing drug combinations to the clinic. For example, recent work identified that Ewing sarcoma tumors are highly sensitive to kt-3283, a bifunctional PARP-HDAC inhibitor (60). PARP proteins respond to DNA damage, including damage caused by DNA replication stress, and we hypothesize that the effects of HDAC inhibitors on DNA replication could contribute to the sensitivity of Ewing sarcoma tumors to dual inhibition of HDAC and PARP (86, 87). Similarly, panobinostat was recently shown to improve the efficacy of standard-of-care drugs, including doxorubicin and etoposide, in targeting Ewing sarcoma cells (61).

HDACs comprise a group of 18 enzymes that are divided into four classes. We found that panobinostat, a pan-HDAC inhibitor, and romidepsin, a class I inhibitor, are both effective in reducing the levels of DNA replication–related proteins (65, 66, 88, 89). Future work will focus on dissecting the contributions of specific HDACs, with an initial focus on the class 1 enzymes (HDAC1, HDAC2, HDAC3, and HDAC8) due to the sensitivity of Ewing sarcoma cell lines to the class 1 HDAC inhibitors romidepsin and entinostat, as well as a published CRISPR screen that identified the sensitivity of Ewing sarcoma cell lines to the loss of class 1 HDACs (59, 61). However, due to the compensatory response of class 1 HDACs to the knockdown of individual HDACs, as well as the correlation between the expression of HDAC1, HDAC2, and HDAC3 and replication genes, we expect that multiple HDACs may contribute to the regulation of the replication genes. In addition, our current work did not distinguish whether HDACs regulate the expression of DNA replication genes by a direct mechanism, via modification of histone acetylation at these specific genes, or an indirect mechanism, such as regulation of upstream transcription factor(s). This question will also be investigated in future work.

Our study focused on the effects of HDAC inhibitors on DNA replication but did not evaluate their impact on Ewing sarcoma tumor growth in vivo using xenograft models. However, previous studies have shown that HDAC inhibitors can reduce the growth of Ewing sarcoma xenografts in mice (59, 63, 90). Future work will assess whether the in vitro mechanisms related to DNA replication observed in this study also apply in vivo. Several HDAC inhibitors—romidepsin, vorinostat, and belinostat—are currently FDA-approved for treating hematologic malignancies such as multiple myeloma and peripheral T-cell lymphoma (91). Panobinostat was previously approved by the FDA for the treatment of multiple myeloma, but this approval was withdrawn due to a lack of completion of postapproval clinical studies. A case report described an adult with progressive, metastatic Ewing sarcoma who was treated with panobinostat and experienced prolonged disease stabilization for 18 months (92). Despite these findings, HDAC inhibitors have shown limited efficacy in solid tumors when used as single agents, prompting the investigation of combination therapies in clinical trials (91). Several early-phase clinical trials have tested HDAC inhibitors in children, mostly as single agents (9399). Notably, vorinostat has been evaluated in combination with temozolomide in children (93, 94). Additionally, the Children’s Oncology Group New Agents for Rhabdomyosarcoma Task Force identified HDAC inhibitors as a priority class of drugs for further investigation (100). For aggressive sarcomas, such as Ewing sarcoma, combination drug therapy will likely be necessary. Understanding how HDAC inhibitors target Ewing sarcoma cells will aid in identifying synergistic drug partners.

In summary, our work demonstrates that HDACs regulate DNA replication and the response to DNA replication stress through multiple mechanisms. We provide evidence that HDAC inhibitors downregulate the expression of multiple components of the pre-RC, including the MCM2–7 proteins and CDT1, as well as the RRM1 and RRM2 subunits of RNR, CHK1, and WEE1. Additionally, we identified that HDAC inhibitors decrease the expression of the BRD4 protein, a bromodomain and extraterminal domain (BET) protein that regulates both the transcriptional program of the EWS::FLI1 oncoprotein and DNA replication. Overall, these results offer novel insights into the mechanism by which HDAC inhibitors target cancer cells and DNA replication.

No disclosures were reported.

S.L. Koppenhafer: Formal analysis, investigation, methodology, writing–review and editing. E.L. Geary: Investigation, methodology, writing–review and editing. M.V. Thomas: Investigation, methodology, writing–review and editing. E.E. Croushore: Investigation. J.A.O. Zimmerman: Formal analysis, writing–review and editing. J.M. Gedminas: Formal analysis, writing–original draft, writing–review and editing. D.E. Quelle: Formal analysis, writing–original draft. R.D. Dodd: Formal analysis, writing–original draft. D.J. Gordon: Conceptualization, resources, formal analysis, supervision, funding acquisition, investigation, methodology, writing–original draft, project administration, writing–review and editing.

D.J. Gordon is supported by the Matt Morrell and Natalie Sanchez Pediatric Cancer Research Foundation, the Stead Fund for Leadership Development & Innovation in Pediatric Medicine, Sammy’s Superheroes, Cal’s Angels, Unravel Pediatric Cancer, Hyundai Hope on Wheels, the Sarah Waul Family Fund, the Charlie Landers Foundation, the Dominic Gentile DVM Sarcoma Research Fund, and NIH Grant R37-CA217910. This work was also supported by a grant (P30CA086862) from the NCI, administered through the Holden Comprehensive Cancer Center at the University of Iowa. The authors would like to acknowledge the use of the University of Iowa Flow Cytometry Facility and the DNA sequencing facility (Genomics Division of the Iowa Institute of Human Genetics), which are supported, in part, by the University of Iowa Carver College of Medicine and the Holden Comprehensive Cancer Center (NCI P30CA086862).

Note: Supplementary data for this article are available at Cancer Research Communications Online (https://aacrjournals.org/cancerrescommun/).

1.
Riggi
N
,
Suvà
ML
,
Stamenkovic
I
.
Ewing’s sarcoma
.
N Engl J Med
2021
;
384
:
154
64
.
2.
Womer
RB
,
West
DC
,
Krailo
MD
,
Dickman
PS
,
Pawel
BR
,
Grier
HE
, et al
.
Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children’s Oncology Group
.
J Clin Oncol
2012
;
30
:
4148
54
.
3.
Martin
JC
,
Hoegel
TJ
,
Lynch
ML
,
Woloszynska
A
,
Melendy
T
,
Ohm
JE
.
Exploiting replication stress as a novel therapeutic intervention
.
Mol Cancer Res
2021
;
19
:
192
206
.
4.
Ubhi
T
,
Brown
GW
.
Exploiting DNA replication stress for cancer treatment
.
Cancer Res
2019
;
79
:
1730
9
.
5.
Kotsantis
P
,
Petermann
E
,
Boulton
SJ
.
Mechanisms of oncogene-induced replication stress: Jigsaw falling into place
.
Cancer Discov
2018
;
8
:
537
55
.
6.
Dobbelstein
M
,
Sørensen
CS
.
Exploiting replicative stress to treat cancer
.
Nat Rev Drug Discov
2015
;
14
:
405
23
.
7.
Berti
M
,
Vindigni
A
.
Replication stress: getting back on track
.
Nat Struct Mol Biol
2016
;
23
:
103
9
.
8.
Su
XA
,
Ma
D
,
Parsons
JV
,
Replogle
JM
,
Amatruda
JF
,
Whittaker
CA
, et al
.
RAD21 is a driver of chromosome 8 gain in Ewing sarcoma to mitigate replication stress
.
Genes Dev
2021
;
35
:
556
72
.
9.
Osgood
CL
,
Tantawy
MN
,
Maloney
N
,
Madaj
ZB
,
Peck
A
,
Boguslawski
E
, et al
.
18F-FLT positron emission tomography (PET) is a pharmacodynamic marker for EWS-FLI1 activity and Ewing sarcoma
.
Sci Rep
2016
;
6
:
33926
.
10.
Tang
S-W
,
Bilke
S
,
Cao
L
,
Murai
J
,
Sousa
FG
,
Yamade
M
, et al
.
SLFN11 is a transcriptional target of EWS-FLI1 and a determinant of drug response in Ewing sarcoma
.
Clin Cancer Res
2015
;
21
:
4184
93
.
11.
Murai
J
,
Thomas
A
,
Miettinen
M
,
Pommier
Y
.
Schlafen 11 (SLFN11), a restriction factor for replicative stress induced by DNA-targeting anti-cancer therapies
.
Pharmacol Ther
2019
;
201
:
94
102
.
12.
Mondal
G
,
Stevers
M
,
Goode
B
,
Ashworth
A
,
Solomon
DA
.
A requirement for STAG2 in replication fork progression creates a targetable synthetic lethality in cohesin-mutant cancers
.
Nat Commun
2019
;
10
:
1686
.
13.
Brohl
AS
,
Solomon
DA
,
Chang
W
,
Wang
J
,
Song
Y
,
Sindiri
S
, et al
.
The genomic landscape of the Ewing Sarcoma family of tumors reveals recurrent STAG2 mutation
.
PLoS Genet
2014
;
10
:
e1004475
.
14.
Crompton
BD
,
Stewart
C
,
Taylor-Weiner
A
,
Alexe
G
,
Kurek
KC
,
Calicchio
ML
, et al
.
The genomic landscape of pediatric Ewing sarcoma
.
Cancer Discov
2014
;
4
:
1326
41
.
15.
Gorthi
A
,
Romero
JC
,
Loranc
E
,
Cao
L
,
Lawrence
LA
,
Goodale
E
, et al
.
EWS-FLI1 increases transcription to cause R-loops and block BRCA1 repair in Ewing sarcoma
.
Nature
2018
;
555
:
387
91
.
16.
Gorthi
A
,
Bishop
AJR
.
Ewing sarcoma fusion oncogene: at the crossroads of transcription and DNA damage response
.
Mol Cell Oncol
2018
;
5
:
e1465014
.
17.
Koppenhafer
SL
,
Goss
KL
,
Terry
WW
,
Gordon
DJ
.
Inhibition of the ATR-CHK1 pathway in Ewing sarcoma cells causes DNA damage and apoptosis via the CDK2-mediated degradation of RRM2
.
Mol Cancer Res
2020
;
18
:
91
104
.
18.
Waters
T
,
Goss
KL
,
Koppenhafer
SL
,
Terry
WW
,
Gordon
DJ
.
Eltrombopag inhibits the proliferation of Ewing sarcoma cells via iron chelation and impaired DNA replication
.
BMC Cancer
2020
;
20
:
1171
.
19.
Goss
KL
,
Koppenhafer
SL
,
Harmoney
KM
,
Terry
WW
,
Gordon
DJ
.
Inhibition of CHK1 sensitizes Ewing sarcoma cells to the ribonucleotide reductase inhibitor gemcitabine
.
Oncotarget
2017
;
8
:
87016
32
.
20.
Goss
KL
,
Gordon
DJ
.
Gene expression signature based screening identifies ribonucleotide reductase as a candidate therapeutic target in Ewing sarcoma
.
Oncotarget
2016
;
7
:
63003
19
.
21.
Koppenhafer
SL
,
Goss
KL
,
Terry
WW
,
Gordon
DJ
.
mTORC1/2 and protein translation regulate levels of CHK1 and the sensitivity to CHK1 inhibitors in Ewing sarcoma cells
.
Mol Cancer Ther
2018
;
17
:
2676
88
.
22.
Nieto-Soler
M
,
Morgado-Palacin
I
,
Lafarga
V
,
Lecona
E
,
Murga
M
,
Callen
E
, et al
.
Efficacy of ATR inhibitors as single agents in Ewing sarcoma
.
Oncotarget
2016
;
7
:
58759
67
.
23.
Lowery
CD
,
Dowless
M
,
Renschler
M
,
Blosser
W
,
VanWye
AB
,
Stephens
JR
, et al
.
Broad spectrum activity of the checkpoint kinase 1 inhibitor prexasertib as a single agent or chemopotentiator across a range of preclinical pediatric tumor models
.
Clin Cancer Res
2019
;
25
:
2278
89
.
24.
Henssen
AG
,
Reed
C
,
Jiang
E
,
Garcia
HD
,
von Stebut
J
,
MacArthur
IC
, et al
.
Therapeutic targeting of PGBD5-induced DNA repair dependency in pediatric solid tumors
.
Sci Transl Med
2017
;
9
:
eaam9078
.
25.
Ohmura
S
,
Marchetto
A
,
Orth
MF
,
Li
J
,
Jabar
S
,
Ranft
A
, et al
.
Translational evidence for RRM2 as a prognostic biomarker and therapeutic target in Ewing sarcoma
.
Mol Cancer
2021
;
20
:
97
.
26.
Martin
JC
,
Sims
JR
,
Gupta
A
,
Hagoel
TJ
,
Gao
L
,
Lynch
ML
, et al
.
CDC7 kinase (DDK) inhibition disrupts DNA replication leading to mitotic catastrophe in Ewing sarcoma
.
Cell Death Discov
2022
;
8
:
85
.
27.
Martin
JC
,
Sims
JR
,
Gupta
A
,
Bakin
AV
,
Ohm
JE
.
WEE1 inhibition augments CDC7 (DDK) inhibitor-induced cell death in Ewing sarcoma by forcing premature mitotic entry and mitotic catastrophe
.
Cancer Res Commun
2022
;
2
:
471
82
.
28.
Sturm
M-J
,
Henao-Restrepo
JA
,
Becker
S
,
Proquitté
H
,
Beck
JF
,
Sonnemann
J
.
Synergistic anticancer activity of combined ATR and ribonucleotide reductase inhibition in Ewing’s sarcoma cells
.
J Cancer Res Clin Oncol
2023
;
149
:
8605
17
.
29.
Jess
J
,
Sorensen
KM
,
Boguslawski
EA
,
Stout
MC
,
Madaj
ZB
,
Caiello
BP
, et al
.
Cell Context is the third axis of synergy for the combination of ATR inhibition and cisplatin in Ewing sarcoma
.
Clin Cancer Res
2024
;
30
:
3533
48
.
30.
Parker
MW
,
Botchan
MR
,
Berger
JM
.
Mechanisms and regulation of DNA replication initiation in eukaryotes
.
Crit Rev Biochem Mol Biol
2017
;
52
:
107
44
.
31.
Kyei Barffour
I
,
Acheampong
DO
.
Prospect of reprogramming replication licensing for cancer drug development
.
Biomed Pharmacother
2021
;
136
:
111190
.
32.
Chuang
C-H
,
Wallace
MD
,
Abratte
C
,
Southard
T
,
Schimenti
JC
.
Incremental genetic perturbations to MCM2-7 expression and subcellular distribution reveal exquisite sensitivity of mice to DNA replication stress
.
PLoS Genet
2010
;
6
:
e1001110
.
33.
Hatoyama
Y
,
Kanemaki
MT
.
The assembly of the MCM2-7 hetero-hexamer and its significance in DNA replication
.
Biochem Soc Trans
2023
;
51
:
1289
95
.
34.
Sun
J
,
Fernandez-Cid
A
,
Riera
A
,
Tognetti
S
,
Yuan
Z
,
Stillman
B
, et al
.
Structural and mechanistic insights into Mcm2-7 double-hexamer assembly and function
.
Genes Dev
2014
;
28
:
2291
303
.
35.
Bryant
VL
,
Elias
RM
,
McCarthy
SM
,
Yeatman
TJ
,
Alexandrow
MG
.
Suppression of reserve MCM complexes chemosensitizes to gemcitabine and 5-fluorouracil
.
Mol Cancer Res
2015
;
13
:
1296
305
.
36.
Ibarra
A
,
Schwob
E
,
Méndez
J
.
Excess MCM proteins protect human cells from replicative stress by licensing backup origins of replication
.
Proc Natl Acad Sci U S A
2008
;
105
:
8956
61
.
37.
Shima
N
,
Pederson
KD
.
Dormant origins as a built-in safeguard in eukaryotic DNA replication against genome instability and disease development
.
DNA Repair (Amst)
2017
;
56
:
166
73
.
38.
Ge
XQ
,
Jackson
DA
,
Blow
JJ
.
Dormant origins licensed by excess Mcm2-7 are required for human cells to survive replicative stress
.
Genes Dev
2007
;
21
:
3331
41
.
39.
Blow
JJ
,
Ge
XQ
,
Jackson
DA
.
How dormant origins promote complete genome replication
.
Trends Biochem Sci
2011
;
36
:
405
14
.
40.
Croushore
EE
,
Koppenhafer
SL
,
Goss
KL
,
Geary
EL
,
Gordon
DJ
.
Activator protein-1 (AP-1) signaling inhibits the growth of Ewing sarcoma cells in response to DNA replication stress
.
Cancer Res Commun
2023
;
3
:
1580
93
.
41.
Mallard
HJ
,
Wan
S
,
Nidhi
P
,
Hanscom-Trofy
YD
,
Mohapatra
B
,
Woods
NT
, et al
.
USP1 expression driven by EWS::FLI1 transcription factor stabilizes Survivin and mitigates replication stress in Ewing sarcoma
.
Mol Cancer Res
2023
;
21
:
1186
204
.
42.
Hingorani
P
,
Dickman
P
,
Garcia-Filion
P
,
White-Collins
A
,
Kolb
EA
,
Azorsa
DO
.
BIRC5 expression is a poor prognostic marker in Ewing sarcoma
.
Pediatr Blood Cancer
2013
;
60
:
35
40
.
43.
Hensel
T
,
Giorgi
C
,
Schmidt
O
,
Calzada-Wack
J
,
Neff
F
,
Buch
T
, et al
.
Targeting the EWS-ETS transcriptional program by BET bromodomain inhibition in Ewing sarcoma
.
Oncotarget
2016
;
7
:
1451
63
.
44.
Gollavilli
PN
,
Pawar
A
,
Wilder-Romans
K
,
Natesan
R
,
Engelke
CG
,
Dommeti
VL
, et al
.
EWS/ETS-driven Ewing sarcoma requires BET bromodomain proteins
.
Cancer Res
2018
;
78
:
4760
73
.
45.
Loganathan
SN
,
Tang
N
,
Fleming
JT
,
Ma
Y
,
Guo
Y
,
Borinstein
SC
, et al
.
BET bromodomain inhibitors suppress EWS-FLI1-dependent transcription and the IGF1 autocrine mechanism in Ewing sarcoma
.
Oncotarget
2016
;
7
:
43504
17
.
46.
Goss
KL
,
Koppenhafer
SL
,
Waters
T
,
Terry
WW
,
Wen
K-K
,
Wu
M
, et al
.
The translational repressor 4E-BP1 regulates RRM2 levels and functions as a tumor suppressor in Ewing sarcoma tumors
.
Oncogene
2021
;
40
:
564
77
.
47.
Koppenhafer
SL
,
Goss
KL
,
Voigt
E
,
Croushore
E
,
Terry
WW
,
Ostergaard
J
, et al
.
Inhibitor of DNA binding 2 (ID2) regulates the expression of developmental genes and tumorigenesis in Ewing sarcoma
.
Oncogene
2022
;
41
:
2873
84
.
48.
Grunewald
TGP
,
Diebold
I
,
Esposito
I
,
Plehm
S
,
Hauer
K
,
Thiel
U
, et al
.
STEAP1 is associated with the invasive and oxidative stress phenotype of Ewing tumors
.
Mol Cancer Res
2012
;
10
:
52
65
.
49.
Richter
GHS
,
Plehm
S
,
Fasan
A
,
Rössler
S
,
Unland
R
,
Bennani-Baiti
IM
, 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
2009
;
106
:
5324
9
.
50.
Ge
SX
,
Jung
D
,
Yao
R
.
ShinyGO: a graphical gene-set enrichment tool for animals and plants
.
Bioinformatics
2020
;
36
:
2628
9
.
51.
Chen
EY
,
Tan
CM
,
Kou
Y
,
Duan
Q
,
Wang
Z
,
Meirelles
GV
, et al
.
Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool
.
BMC Bioinformatics
2013
;
14
:
128
.
52.
Ge
SX
,
Son
EW
,
Yao
R
.
iDEP: an integrated web application for differential expression and pathway analysis of RNA-Seq data
.
BMC Bioinformatics
2018
;
19
:
534
.
53.
Croushore
EE
,
Stipp
CS
,
Gordon
DJ
.
EWS-FLI1 and activator protein-1 (AP-1) reciprocally regulate extracellular-matrix proteins in Ewing sarcoma cells
.
Int J Mol Sci
2024
;
25
:
8595
.
54.
Tsherniak
A
,
Vazquez
F
,
Montgomery
PG
,
Weir
BA
,
Kryukov
G
,
Cowley
GS
, et al
.
Defining a cancer dependency map
.
Cell
2017
;
170
:
564
76.e16
.
55.
Dharia
NV
,
Kugener
G
,
Guenther
LM
,
Malone
CF
,
Durbin
AD
,
Hong
AL
, et al
.
A first-generation pediatric cancer dependency map
.
Nat Genet
2021
;
53
:
529
38
.
56.
Savola
S
,
Klami
A
,
Myllykangas
S
,
Manara
C
,
Scotlandi
K
,
Picci
P
, et al
.
High expression of complement component 5 (C5) at tumor site associates with superior survival in Ewing’s sarcoma family of tumour patients
.
ISRN Oncol
2011
;
2011
:
168712
.
57.
Li
X
,
Su
Y
,
Madlambayan
G
,
Edwards
H
,
Polin
L
,
Kushner
J
, et al
.
Antileukemic activity and mechanism of action of the novel PI3K and histone deacetylase dual inhibitor CUDC-907 in acute myeloid leukemia
.
Haematologica
2019
;
104
:
2225
40
.
58.
Li
X
,
Su
Y
,
Hege
K
,
Madlambayan
G
,
Edwards
H
,
Knight
T
, et al
.
The HDAC and PI3K dual inhibitor CUDC-907 synergistically enhances the antileukemic activity of venetoclax in preclinical models of acute myeloid leukemia
.
Haematologica
2021
;
106
:
1262
77
.
59.
Schmidt
O
,
Nehls
N
,
Prexler
C
,
von Heyking
K
,
Groll
T
,
Pardon
K
, et al
.
Class I histone deacetylases (HDAC) critically contribute to Ewing sarcoma pathogenesis
.
J Exp Clin Cancer Res
2021
;
40
:
322
.
60.
Ramos
L
,
Truong
S
,
Zhai
B
,
Joshi
J
,
Ghaidi
F
,
Lizardo
MM
, et al
.
A bifunctional PARP-HDAC inhibitor with activity in Ewing sarcoma
.
Clin Cancer Res
2023
;
29
:
3541
53
.
61.
Smith
KH
,
Trovillion
EM
,
Sholler
C
,
Gandra
D
,
McKinney
KQ
,
Mulama
D
, et al
.
Panobinostat synergizes with chemotherapeutic agents and improves efficacy of standard-of-care chemotherapy combinations in Ewing sarcoma cells
.
Cancers (Basel)
2024
;
16
:
3565
.
62.
Souza
BK
,
da Costa Lopez
PL
,
Menegotto
PR
,
Vieira
IA
,
Kersting
N
,
Abujamra
AL
, et al
.
Targeting histone deacetylase activity to arrest cell growth and promote neural differentiation in Ewing sarcoma
.
Mol Neurobiol
2018
;
55
:
7242
58
.
63.
El-Naggar
AM
,
Somasekharan
SP
,
Wang
Y
,
Cheng
H
,
Negri
GL
,
Pan
M
, et al
.
Class I HDAC inhibitors enhance YB-1 acetylation and oxidative stress to block sarcoma metastasis
.
EMBO Rep
2019
;
20
:
e48375
.
64.
Sakimura
R
,
Tanaka
K
,
Nakatani
F
,
Matsunobu
T
,
Li
X
,
Hanada
M
, et al
.
Antitumor effects of histone deacetylase inhibitor on Ewing’s family tumors
.
Int J Cancer
2005
;
116
:
784
92
.
65.
Li
G
,
Tian
Y
,
Zhu
W-G
.
The roles of histone deacetylases and their inhibitors in cancer therapy
.
Front Cell Dev Biol
2020
;
8
:
576946
.
66.
Falkenberg
KJ
,
Johnstone
RW
.
Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders
.
Nat Rev Drug Discov
2014
;
13
:
673
91
.
67.
Gallinari
P
,
Di Marco
S
,
Jones
P
,
Pallaoro
M
,
Steinkühler
C
.
HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics
.
Cell Res
2007
;
17
:
195
211
.
68.
Lyu
K
,
Kumagai
A
,
Dunphy
WG
.
RPA-coated single-stranded DNA promotes the ETAA1-dependent activation of ATR
.
Cell Cycle
2019
;
18
:
898
913
.
69.
Wang
L
,
Liu
X
.
Pan-cancer multi-omics analysis of minichromosome maintenance proteins (MCMs) expression in human cancers
.
Front Biosci (Landmark Ed)
2023
;
28
:
230
.
70.
Radhakrishnan
A
,
Gangopadhyay
R
,
Sharma
C
,
Kapardar
RK
,
Sharma
NK
,
Srivastav
R
.
Unwinding helicase MCM functionality for diagnosis and therapeutics of replication abnormalities associated with cancer: a review
.
Mol Diagn Ther
2024
;
28
:
249
64
.
71.
Pattenden
SG
,
Simon
JM
,
Wali
A
,
Jayakody
CN
,
Troutman
J
,
McFadden
AW
, et al
.
High-throughput small molecule screen identifies inhibitors of aberrant chromatin accessibility
.
Proc Natl Acad Sci U S A
2016
;
113
:
3018
23
.
72.
Bharathy
N
,
Berlow
NE
,
Wang
E
,
Abraham
J
,
Settelmeyer
TP
,
Hooper
JE
, et al
.
Preclinical rationale for entinostat in embryonal rhabdomyosarcoma
.
Skelet Muscle
2019
;
9
:
12
.
73.
Donati
B
,
Lorenzini
E
,
Ciarrocchi
A
.
BRD4 and cancer: going beyond transcriptional regulation
.
Mol Cancer
2018
;
17
:
164
.
74.
Richter
GHS
,
Hensel
T
,
Schmidt
O
,
Saratov
V
,
von Heyking
K
,
Becker-Dettling
F
, et al
.
Combined inhibition of epigenetic readers and transcription initiation targets the EWS-ETS transcriptional program in Ewing sarcoma
.
Cancers (Basel)
2020
;
12
:
304
.
75.
Zhang
J
,
Dulak
AM
,
Hattersley
MM
,
Willis
BS
,
Nikkilä
J
,
Wang
A
, et al
.
BRD4 facilitates replication stress-induced DNA damage response
.
Oncogene
2018
;
37
:
3763
77
.
76.
Lam
FC
,
Kong
YW
,
Huang
Q
,
Vu Han
T-L
,
Maffa
AD
,
Kasper
EM
, et al
.
BRD4 prevents the accumulation of R-loops and protects against transcription-replication collision events and DNA damage
.
Nat Commun
2020
;
11
:
4083
.
77.
Edwards
DS
,
Maganti
R
,
Tanksley
JP
,
Luo
J
,
Park
JJH
,
Balkanska-Sinclair
E
, et al
.
BRD4 prevents R-loop formation and transcription-replication conflicts by ensuring efficient transcription elongation
.
Cell Rep
2020
;
32
:
108166
.
78.
Sollazzo
MR
,
Benassi
MS
,
Magagnoli
G
,
Gamberi
G
,
Molendini
L
,
Ragazzini
P
, et al
.
Increased c-myc oncogene expression in Ewing’s sarcoma: correlation with Ki67 proliferation index
.
Tumori
1999
;
85
:
167
73
.
79.
Kawano
M
,
Tanaka
K
,
Itonaga
I
,
Iwasaki
T
,
Tsumura
H
.
c-Myc represses tumor-suppressive microRNAs, let-7a, miR-16 and miR-29b, and induces cyclin D2-mediated cell proliferation in Ewing’s sarcoma cell line
.
PLoS One
2015
;
10
:
e0138560
.
80.
Mahmoud
AM
,
Zekri
W
,
Khorshed
EN
,
Shalaby
LM
.
Prognostic significance of survivin expression in pediatric Ewing sarcoma
.
Pediatr Hematol Oncol
2022
;
39
:
16
27
.
81.
Grünewald
TGP
,
Cidre-Aranaz
F
,
Surdez
D
,
Tomazou
EM
,
de Álava
E
,
Kovar
H
, et al
.
Ewing sarcoma
.
Nat Rev Dis Primers
2018
;
4
:
5
.
82.
Kovar
H
.
Blocking the road, stopping the engine or killing the driver? Advances in targeting EWS/FLI-1 fusion in Ewing sarcoma as novel therapy
.
Expert Opin Ther Targets
2014
;
18
:
1315
28
.
83.
Xiang
S
,
Craig
KC
,
Luo
X
,
Welch
DL
,
Ferreira
RB
,
Lawrence
HR
, et al
.
Identification of ATP-competitive human CMG helicase inhibitors for cancer intervention that disrupt CMG-replisome function
.
Mol Cancer Ther
2024
;
23
:
1568
85
.
84.
Slaughter
MJ
,
Shanle
EK
,
Khan
A
,
Chua
KF
,
Hong
T
,
Boxer
LD
, et al
.
HDAC inhibition results in widespread alteration of the histone acetylation landscape and BRD4 targeting to gene bodies
.
Cell Rep
2021
;
34
:
108638
.
85.
Chang
T-Y
,
Yan
Y
,
Yu
Z-Y
,
Rathore
M
,
Lee
N-Z
,
Tseng
H-J
, et al
.
Combined HDAC8 and checkpoint kinase inhibition induces tumor-selective synthetic lethality in preclinical models
.
J Clin Invest
2024
;
134
:
e165448
.
86.
Bryant
HE
,
Petermann
E
,
Schultz
N
,
Jemth
A-S
,
Loseva
O
,
Issaeva
N
, et al
.
PARP is activated at stalled forks to mediate Mre11-dependent replication restart and recombination
.
EMBO J
2009
;
28
:
2601
15
.
87.
Cong
K
,
Peng
M
,
Kousholt
AN
,
Lee
WTC
,
Lee
S
,
Nayak
S
, et al
.
Replication gaps are a key determinant of PARP inhibitor synthetic lethality with BRCA deficiency
.
Mol Cell
2021
;
81
:
3128
44.e7
.
88.
Park
S-Y
,
Kim
J-S
.
A short guide to histone deacetylases including recent progress on class II enzymes
.
Exp Mol Med
2020
;
52
:
204
12
.
89.
Seto
E
,
Yoshida
M
.
Erasers of histone acetylation: the histone deacetylase enzymes
.
Cold Spring Harb Perspect Biol
2014
;
6
:
a018713
.
90.
Jaboin
J
,
Wild
J
,
Hamidi
H
,
Khanna
C
,
Kim
CJ
,
Robey
R
, et al
.
MS-27-275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumors
.
Cancer Res
2002
;
62
:
6108
15
.
91.
Shi
M-Q
,
Xu
Y
,
Fu
X
,
Pan
D-S
,
Lu
X-P
,
Xiao
Y
, et al
.
Advances in targeting histone deacetylase for treatment of solid tumors
.
J Hematol Oncol
2024
;
17
:
37
.
92.
van Maldegem
AM
,
Bovée
JVMG
,
Gelderblom
H
.
Panobinostat-A potential treatment for metastasized Ewing sarcoma? A case report
.
Pediatr Blood Cancer
2016
;
63
:
1840
3
.
93.
Peters
KB
,
Lipp
ES
,
Miller
E
,
Herndon
JE
,
McSherry
F
,
Desjardins
A
, et al
.
Phase I/II trial of vorinostat, bevacizumab, and daily temozolomide for recurrent malignant gliomas
.
J Neurooncol
2018
;
137
:
349
56
.
94.
Lee
EQ
,
Puduvalli
VK
,
Reid
JM
,
Kuhn
JG
,
Lamborn
KR
,
Cloughesy
TF
, et al
.
Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03
.
Clin Cancer Res
2012
;
18
:
6032
9
.
95.
Witt
O
,
Milde
T
,
Deubzer
HE
,
Oehme
I
,
Witt
R
,
Kulozik
A
, et al
.
Phase I/II intra-patient dose escalation study of vorinostat in children with relapsed solid tumor, lymphoma or leukemia
.
Klin Padiatr
2012
;
224
:
398
403
.
96.
Hummel
TR
,
Wagner
L
,
Ahern
C
,
Fouladi
M
,
Reid
JM
,
McGovern
RM
, et al
.
A pediatric phase 1 trial of vorinostat and temozolomide in relapsed or refractory primary brain or spinal cord tumors: a Children’s Oncology Group phase 1 consortium study
.
Pediatr Blood Cancer
2013
;
60
:
1452
7
.
97.
Zorzi
AP
,
Bernstein
M
,
Samson
Y
,
Wall
DA
,
Desai
S
,
Nicksy
D
, et al
.
A phase I study of histone deacetylase inhibitor, pracinostat (SB939), in pediatric patients with refractory solid tumors: IND203 a trial of the NCIC IND program/C17 pediatric phase I consortium
.
Pediatr Blood Cancer
2013
;
60
:
1868
74
.
98.
van Tilburg
CM
,
Milde
T
,
Witt
R
,
Ecker
J
,
Hielscher
T
,
Seitz
A
, et al
.
Phase I/II intra-patient dose escalation study of vorinostat in children with relapsed solid tumor, lymphoma, or leukemia
.
Clin Epigenetics
2019
;
11
:
188
.
99.
Bukowinski
A
,
Chang
B
,
Reid
JM
,
Liu
X
,
Minard
CG
,
Trepel
JB
, et al
.
A phase 1 study of entinostat in children and adolescents with recurrent or refractory solid tumors, including CNS tumors: trial ADVL1513, Pediatric Early Phase-Clinical Trial Network (PEP-CTN)
.
Pediatr Blood Cancer
2021
;
68
:
e28892
.
100.
Pacenta
HL
,
Allen-Rhoades
W
,
Langenau
D
,
Houghton
PJ
,
Keller
C
,
Heske
CM
, et al
.
Prioritization of novel agents for patients with rhabdomyosarcoma: a report from the Children’s Oncology group (COG) new agents for rhabdomyosarcoma task force
.
J Clin Med
2021
;
10
:
1416
.
This open access article is distributed under the Creative Commons Attribution 4.0 International (CC BY 4.0) license.

Supplementary data