Small Molecule Screen Identifies Inhibitor of PAX3-FOXO1 Activity
A small molecule library of 62,643 pure compounds (NCI Molecular Targets Program) was screened at a dose of 10 µM for 24-hours using a genetically engineered RH4 FP-RMS cell line expressing a PAX3-FOXO1-selective ALK super enhancer-driven luciferase 9. In parallel, we also tested the compounds on an RH4 cell line with a constitutively active CMV promoter-driven luciferase to monitor general transcriptional inhibition. We selected molecules with two characteristics: 1) Selective decrease in ALK super enhancer-driven luciferase relative to CMV promoter-driven luciferase; and 2) lack of non-specific toxicity at this early timepoint as measured by XTT cell viability assay (Fig. 1A). As described previously, we integrated the ALK luciferase, CMV luciferase, and XTT assays to generate a weighted average score that resulted in the identification of 573 hit molecules 7. Dose response data for these molecules was obtained in a follow-up screen consisting of five 10-fold dilutions ranging from 20 µM to 2 nM. This further reduced the candidate small molecules to 64 (Supplementary Table 1, Table 1), which were classified into 3 major groups 7. The first group showed decreased ALK super enhancer-driven luciferase concomitant with increased CMV-driven luciferase as observed with histone deacetylase inhibitors (HDACi) exemplified by N1302 (1-alaninechlamydocin) 7, which we designated HDACi-like compounds. A second group showed decreased ALK super-enhancer-driven luciferase without a significant change in CMV-driven luciferase, which we referred to as ALK-selective compounds. Lastly, compounds that strongly decreased both ALK super-enhancer and CMV-driven luciferase below 10% of control were denoted strong inhibitors (Fig. 1B). Besides N1302, there were 24 other compounds with known mechanisms of action including Midostaurin (PKC-412), a previously identified inhibitor of PAX3-FOXO1, which validated our screening methodology 10.
To investigate potential mechanisms of action of novel compounds, we manually selected few inhibitors from each group; HDAC-like, ALK-selective, and strong inhibitor for RNA-seq analysis following treatment of RH4 FP-RMS cells (Fig. 1B). Gene set enrichment analysis (GSEA) 11, 12 showed that PFI-63 (UPCMLD0ENAT5834780) treatment most closely phenocopied shRNA knockdown of PAX3-FOXO1 when looking at the sum of normalized enrichment scores of downregulated PAX3-FOXO1 gene sets. (Fig. 1C, Supplementary Fig. 1A and Table 2) 9, 13. In addition, myogenesis and apoptosis gene sets were significantly enriched, suggesting that PFI-63 may induce FP-RMS differentiation and, ultimately, apoptosis. In line with this, western analysis demonstrated increased MYOG levels and PARP cleavage compared to DMSO control (Fig. 1D and Supplementary Fig. 1B). Direct enzymatic assay confirmed that caspase 3/7 activities were significantly increased (Supplementary Fig. 1C). Notably, PFI-63 action did not directly decrease the amount of PAX3-FOXO1 protein (Fig. 1D).
The half-maximal inhibitory concentrations (IC50) of PFI-63 on FP-RMS cell lines RH4, RH30, and SCMC were in the single digit µM range (Fig. 1E). Next, we tested other pediatric solid cancer cell lines to determine broader applicability of PFI-63. IC50 values for PFI-63 on fusion negative RMS cell lines (RD: 7.16 µM and CTR: 2.92 µM), Ewing’s sarcoma (TC-32: 1.7 µM and A673: 3.7 µM) and osteosarcoma (OSA: 7.5 µM and HU09: 3.1 µM) were also in the single digit µM range (Supplementary Fig. 1D). We also tested PFI-63 on the NCI-60 cancer cell line panel 14, which showed that PFI-63 was mostly cytostatic in adult cancer cell lines (Supplementary Fig. 2). Given that PFI-63 downregulated PAX3-FOXO1 target gene expression in FP-RMS and had broad activity in other pediatric solid tumor cell lines, we next wanted to determine its molecular target(s).
PFI-63 Targets Histone Lysine Demethylases
First, we searched for other gene expression signatures that were significantly enriched following treatment with PFI-63 11, 15. Remarkably, among the most highly upregulated gene sets by PFI-63 were genes associated with GSK-J4 and JIB-04 treatment (Fig. 1F) 16, 17. GSK-J4 and JIB-04 are inhibitors of the Jumonji domain (JmjC) family of histone lysine demethylases (KDMs) that are involved in epigenetic control of transcription. This suggested that PFI-63 may act by inhibiting KDMs. In support of this notion, submission of the PFI-63 chemical structure to the SEA (Similarity Ensemble Approach) Search Server (https://sea.bkslab.org/) to generate predictions for potential molecular targets revealed that PFI-63 could be a ligand for multiple JmjC KDMs (Supplementary Table S1, Table 2) 18, 19. Moreover, ATAC-seq after treatment with PFI-63 to assess new open areas of chromatin showed enrichment of a gene set induced by treatment with the KDM1A/LSD1 inhibitor SP2509 (Supplementary Fig. 3A) 20. Therefore, we hypothesized that PFI-63’s mechanism of action to be the inhibition of KDMs. Hence, we assessed the direct inhibition of KDMs by PFI-63 using cell free in vitro enzyme inhibition assays. Confirming our hypothesis, PFI-63 inhibited multiple KDMs including KDM3B, KDM4B, KDM5A, and KDM6B, with highest potency against KDM3B at an IC50 of 7 µM (Fig. 1G). PyMOL alignment of PFI-63 to the enzyme pocket of KDM3B is shown in Fig. 1H. In contrast, PFI-63 had no significant inhibitory activity against HDAC1, HDAC2, HDAC3, or PRMT5 up to a concentration of 10 µM. Given these findings, we performed western analysis of PFI-63-treated FP-RMS cell lines for changes in methylation status of histone 3 lysines: H3K9 (demethylated by KDM3B, KDM4B, KDM1A), H3K4 (KDM1A, KDM5A) and H3K27 (KDM6B). After 24-hour treatment of RH4 and RH30 cells with PFI-63, we found significant increases in the methylation of all three lysine substrates compared to DMSO treatment, confirming that multiple histone demethylases are targeted by PFI-63 in FP-RMS cells (Fig. 1I and Supplementary Fig. 3B). Given the novel semi-selectivity of PFI-63 towards KDM3B, we next investigated KDM3B expression levels in RMS cell lines using the Broad Institute’s Dependency Map (DepMap) portal. We observed that RMS cell lines had the second highest average levels of expression of KDM3B compared to other cancer lines, indicating that KDM3B is significantly expressed in RMS and a potential target for therapy (Supplementary Fig. 3C).
Identification of Chemical Analog of PFI-63 with improved drug-like properties
PFI-63 had reduced solubility in water with a partition coefficient logP of 2.36 and thus has poor drug-like properties and makes preclinical in vivo testing a challenge. Hence, we conducted a molecular similarity search with PFI-63 in the Enamine catalog and identified 26 commercially available compounds of similar structure (Supplementary Table 1, Table 3). Performing the ALK super enhancer-driven luciferase screen with the 26 compounds identified one new compound designated as PFI-90 (Compound 26) (Fig. 2A), which showed significant inhibition of PAX3-FOXO1 driven luciferase (Supplementary Table 1, Table 3). PFI-90 had increased water solubility with a partition coefficient logP of 0.82 and showed low to sub-micromolar IC50 activity in FP-RMS (Fig. 2B). PFI-90 was also tested on a large panel of adult cancer cell lines (NCI-60) which again showed mostly non-cytotoxic growth inhibitory effects except for renal cancer cell lines where cell killing was observed at the highest dose tested (Supplementary Fig. 4).
Similar to PFI-63, cell free in vitro enzyme inhibition assays confirmed that PFI-90 had highest inhibitory selectivity towards KDM3B (Fig. 2C). Western analysis showed an increase of H3K4me3 and H3K9me2 but only mild increase of H3K27me3 at doses up to 4µM (Fig. 2D and Supplementary Fig. 5A). This was consistent with PFI-90 having higher potency for KDM3B, KDM4B, KDM5A, and KDM1A which target H3K9 (KDM3B, KDM4B, KDM1A) and H3K4 (KDM1A, KDM5A) than KDM6B (H3K27). RNA-seq analysis at two time points, 6 hours and 24 hours after PFI-90 treatment showed significant enrichment of the myogenesis gene set at the 6-hour time point without enrichment of the apoptosis gene set (Fig. 2E and Supplementary Table 3, Table 1). However, at the 24-hour time point, myogenesis was no longer enriched and there was an enrichment of the apoptosis gene set along with downregulation of PAX3-FOXO1 targets (Supplementary Table 3, Table 2). Western analysis showed an increase of MYOG at 6 and 12 hours with a sharp decrease at 24 hours consistent with the GSEA analysis (Fig. 2F and Supplementary Fig. 5B). Additionally, there was initiation of PARP cleavage at 24 hours indicating apoptosis which was consistent with the GSEA result at the 24-hour time point. We also quantitated caspase 3/7 for apoptosis and found robust increase in caspase after PFI-90 treatment (Supplementary Fig. 5C). Lastly, we examined the transcript levels of the KDMs after treatment of PFI-63 and PFI-90 and found no significant change compared to DMSO treatment, indicating that the biological effect is due to enzymatic inhibition and not due to downregulation of KDM transcription (Supplementary Fig. 5D).
Biophysical Interaction Between PFI-90 and KDM3B
To establish that PFI-90 directly binds to KDM3B, we performed a series of bio-orthogonal ligand-binding detection experiments using nuclear magnetic resonance (NMR) techniques. For the NMR experiments, we employed the truncated KDM3B protein previously used to determine the crystal structure of the KDM3B enzymatic pocket 21. We initially obtained standard proton (1H), carbon (13C), and HSQC NMR spectra of samples containing PFI-90 and 2-OG alone and together in buffer, as well as in the presence or absence of the KDM3B construct (Supplementary Fig. 6A). These results were used as reference for ligand-observed NMR experiments, allowing for unambiguous assignment of peaks to the compound structure. We then subjected these samples to Water-Ligand Observed via Gradient Spectroscopy (WaterLOGSY) 1H NMR. We observed positive proton peaks for PFI-90 only in the presence of KDM3B indicating protein binding (Fig. 2G). When KDM3B was absent, PFI-90 had negative peaks indicating lack of binding as expected. Our non-binding negative control compound N-Methyl-L-Valine (NMV) showed negative peaks both with and without KDM3B indicating no binding as expected.
Next, we subjected the samples to Carr-Purcell-Meiboom-Gill Relaxation-Editing 1H NMR (CPMG) to confirm binding using an orthogonal NMR method. CPMG analysis of PFI-90 showed binding of PFI-90 to KDM3B indicated by the attenuation in the PFI-90 peaks when compared to PFI-90 without KDM3B (Fig. 2H). Again, our non-binding negative control molecule NMV showed that there was no binding by CPMG as indicated by lack of attenuation of NMV peaks in the presence of KDM3B. Interestingly, nearly all 1H signals of PFI-90 showed attenuation to ~ 75%, indicating that the vast majority of the PFI-90 structure binds to the enzymatic portion of the KDM3B protein in solution.
Finally, we also used Surface Plasmon Resonance (SPR) to determine binding of PFI-90 to synthesized full length KDM3B which showed robust enzymatic activity in demethylating H3K9me2 (Supplementary Fig. 6B). SPR demonstrated binding between PFI-90 to KDM3B with a Kd of 7.68 x10− 6 M (Fig. 2I). Hence, we show, using 3 different methods that PFI-90 biophysically binds to KDM3B.
KDM3B is Essential for Survival in FP-RMS
Given that our novel compound targets KDM3B, we investigated whether KDM3B was essential for survival of FP-RMS. We developed a CRISPR library with sgRNA designed across all KDM3B exons. As controls, we used sgRNA for 8 essential genes including PAX3-FOXO1, MYOD, and 20 non-targeting sgRNA from the Brunello CRISPR library 22. We found that 7.1% of sgRNAs which target KDM3B were depleted by > 4-fold in the RH4 FP-RMS cell line with hot spots in the enzymatic domain of KDM3B, indicating that it is in fact essential for survival of FP-RMS (Fig. 3A). It is noteworthy that KDM3B has not previously been recognized as a dependent gene in FP-RMS (e.g., in DepMap), most likely due to a limited selection of sgRNAs against this gene.
KDM Knockdown Recapitulates the PFI-90-induced Transcriptome
To demonstrate that PFI-90 downregulation of PAX3-FOXO1 target genes is through the inhibition of KDMs especially KDM3B, we knocked down the top 4 KDMs inhibited by PFI-90, namely KDM3B, KDM1A, KDM4B, and KDM5A using CRISPRi. We achieved effective knockdown of KDMs measured at the transcript level and at the protein level (Fig. 3B-C and Supplementary Fig. 7A-C). RNA-seq analysis followed by GSEA enrichment showed that KDM3B knockdown significantly downregulated all 5 PAX3-FOXO1-related gene sets that were downregulated by PFI-90 (Fig. 3D and Supplementary Table 3, Table 3–10). Comparison of KDM knockdown to PAX3-FOXO1 knockdown using shRNA showed that KDM3B knockdown decreased PAX3-FOXO1 gene sets most robustly (Supplementary Fig. 7D). However, we did not see enrichment of the upregulated gene sets observed following PFI-90 treatment, namely, those involved in myogenesis or apoptosis. Given that KDM3B removes transcriptionally repressive methyl groups from H3K9, enrichment of the same downregulated gene sets was in line with inhibition of KDM3B function. We reasoned that since H3K4 methylation was also increased and since methylated H3K4 is a marker of transcriptional activation, we investigated upregulated GSEA gene sets of KDM1A and KDM5A knockdowns, both of which target H3K4. Interestingly, we found enrichment for myogenesis and apoptosis following KDM1A knockdown but not in KDM5A knockdown cells. KDM1A knockdown also downregulated PAX3-FOXO1 gene sets although not as robustly as KDM3B knockdown. The combined knockdown of KDM1A and KDM3B showed enrichment for myogenic differentiation and apoptosis gene sets together with downregulation of PAX3-FOXO1 targets, most closely recapitulating the PFI-90-induced transcriptome in RH4 cells (Fig. 3D).
scRNA-seq Shows PFI-90-Induced Decrease PAX3-FOXO1 Signature, Myoblast Signature, and G2/M Population
In order to study the effects of PFI-90 at the single-cell level, we performed scRNA-seq analysis after treating cells with PFI-90 for 16 hours, a time point before apoptosis. Integrating the PFI-90 and DMSO samples together, Cluster 2 showed the largest fold difference in cell number between the PFI-90 and DMSO groups with 82.0% of the cluster comprised of DMSO treated cells (Fig. 4A-B). Cluster 2 was also the largest cluster of DMSO treated cells overall and hence represented a major population of RH4 cells during exponential growth. Clusters 3 and 8 showed the greatest fold change following PFI-90 treatment compared to DMSO. To characterize the clusters, we examined myogenic differentiation using the top 50 differentially expressed genes from a previously published scRNA-seq profile (Supplementary Table 4, Table 1–2) 23. RMS cells are actively dividing in culture, resembling highly proliferative myoblast cells. Interestingly, myoblast signature for Cluster 2 was significantly higher than Clusters 1, 3, 6, and 8 (Fig. 4C and Supplementary Table 4, Table 3). For the less proliferative myocyte signature, Cluster 6 had significantly higher myocytes signature compared to the rest of the clusters. Differential gene expression analysis showed Cluster 6 with high expression of skeletal muscle genes including multiple myosin light chain and heavy chain genes (Supplementary Table 4, Table 4–11). Clusters 3 and 8 which increased the most after PFI-90 treatment had reduced expression of myoblast genes indicating that PFI-90 treatment led to an increase in the number of cells with the lowest myoblast signature. When we examined the PAX3-FOXO1 signature 8, we found that Clusters 3 and 8 also had lower expression of PAX3-FOXO1 downstream targets. Heatmap hierarchical clustering using leading edge genes from myoblast signature and PAX3-FOXO1 signature both showed that Clusters 3 and 8 separated from the rest of the Clusters (Supplementary Fig. 8A). These findings demonstrated a strong correlation between PAX3-FOXO1 expression and maintenance of the myoblast signature. We next performed cell cycle analysis of the scRNA-seq clusters (Fig. 4D-E). Overall, there was an increase of cells in G1 and S while G2M was decreased after PFI-90 treatment indicating potential G2M arrest. When examining which clusters were associated with the decrease in G2M, we saw significant decrease in Cluster 2 after PFI-90 treatment (Fig. 4E). To better understand the cell cycle changes, we performed flow cytometry analysis at 24 hours after treatment with PFI-90 vs DMSO in SCMC cell line. We found profound accumulation of S phase cells while G2 and M was significantly decreased indicating a S phase blockade (Fig. 4F). These findings from scRNA-seq indicated that PFI-90 treatment resulted in decreased expression of PAX3-FOXO1 downstream targets concomitant with decreased myoblast signature and cell division in individual cells.
PFI-90 Increases H3K4 and H3K9 Methylation
To determine how inhibition of KDM3B results in abrogation of PAX3-FOXO1 action, we performed ChIP-seq analysis using antibodies against PAX3-FOXO1, H3K4me3, H3K9me2, H3K27me3, and H3K27ac. First, we performed ChromHMM analysis using our previously published ChIP-seq data to assign the chromatin into 16 states 8, 9. We observed that H3K9me2 was increased throughout the entire genome after treatment with PFI-90 compared to DMSO, with the most pronounced increase at enhancer regions (Fig. 5A). ChromHMM also showed that H3K4me3 was increased mostly in the promoter region which is consistent with its function as a transcription initiation mark. Next, we looked at the transcription start sites (TSS) and PAX3-FOXO1 binding sites. We found that there was an increase in histone methylation at H3K9me2 both at the TSS and PAX3-FOXO1 binding sites in PFI-90 treated cells compared to DMSO (Fig. 5B). This was consistent with PFI-90 having highest inhibition of KDM3B which demethylates histones at H3K9. There was also an increase in H3K4me3 around TSS following treatment with PFI-90, consistent with PFI-90 inhibiting KDM1A and KDM5A which demethylate H3K4. We saw no changes in the methylation status of H3K27. These changes corresponded with PFI-90 selectivity for each of the respective histone marks.
Since PAX3-FOXO1 drives core regulatory transcription factors at super enhancer sites marked with H3K27ac, we looked to see if there were any changes to enhancer-related histone marks around the PAX3-FOXO1 binding sites. There was no change in H3K27ac at PAX3-FOXO1 sites with treatment of PFI-90 compared to DMSO (Fig. 5B). Super enhancer ROSE analysis also showed no changes in super enhancers when RH4 cells were treated with PFI-90 compared to DMSO (Supplementary Fig. 9A). PFI-90 also did not change the binding of PAX3-FOXO1 to target chromatin regions as assessed by ChromHMM despite increases in H3K9me2, consistent with a previous report of PAX3-FOXO1 having the ability to bind heterochromatin as a pioneering factor 24. We also examined the protein level of PAX3-FOXO1 by Western since PAX3-FOXO1 binds super enhancers and found that PFI-90 did not directly decrease PAX3-FOXO1 protein levels (Supplementary Fig. 9B). These results are similar to PFI-63 and consistent with the ChIP-seq data (Supplementary Fig. 9C).
Given that H3K9me2 was increased most significantly at enhancer regions, we wanted to know the relative distribution of H3K9me2 at PAX3-FOXO1 binding sites compared to other transcription factors. Focusing on enhancer regions associated with genes based on RNA-seq transcript level using EDEN, we selected the top 10% with increased H3K9me2 after PFI-90 treatment and performed ChIP Enrichment Analysis (ChEA) via Enricher which compares gene lists with previously published ChIP-seq of transcription factors (Fig. 5C and Supplementary Table 5, Table 1)25. PAX3-FOXO1 was the top significant transcription factor overlapping with H3K9me2 followed by ZNF217 which is one of the PAX3-FOXO1 core regulatory transcription factors 8. Next, we looked to see if the increase in H3K4me3 methylation at promoter regions was associated with enrichment of biological processes predicted by RNA-seq GSEA, namely myogenesis and apoptosis. We detected new H3K4me3 peaks after treatment with PFI-90 and performed gene ontology analysis which showed strong (8.9-fold) enrichment for muscle differentiation as well as 3.4 fold enrichment for apoptosis (Fig. 5D and Supplementary Table 5, Table 2) 26. Therefore, these ChIP analysis results validated the bulk RNA-seq data; i.e., PFI-90 suppresses PAX3-FOXO1 function and upregulates myogenesis as well as apoptosis programs.
PFI-90 Decreases Chromatin Loop Strength and Decreases Topologically Associating Domains (TADs) at PAX3-FOXO1 Sites
Given that there were no changes to PAX3-FOXO1, H3K27me3, and H3K27ac binding and respective super enhancer calls following treatment with PFI-90, we asked if the increase in H3K9me2 and H3K4me3 is sufficient to change the chromatin structure in FP-RMS cells. We performed Hi-C and assessed if PFI-90 treatment was associated with decreased number of loops given that H3K9me2 is a repressive heterochromatin mark. We found that there were minimal changes in the number of loops (675 loss and 582 gain). Genes associated with these loops did not show any statistically significant association with gene sets. However, we found that although there was no difference in loop number, there was decrease in loop strength (Supplementary Fig. 10A-B). Next, we looked to see if TADs were changed by PFI-90 given the decrease in loop strength. Interestingly, there was significant decrease in TAD score in PFI-90 treated cells compared to DMSO (Supplementary Fig. 10C). Genes associated with the decreased TADs and loop strength were enriched for PAX3-FOXO1 target genes by GSEA gene sets (Supplementary Table 5, Table 3). Decrease in TADs and loop strength at PAX3-FOXO1 sites suggests that decrease in PAX3-FOXO1 target transcripts, observed by RNA-seq, may be related to the increase in H3K9me2 at PAX3-FOXO1 sites.
PFI-90 Decreased RNA Pol2 Binding and Phosphorylation
RNA Pol2 binding and phosphorylation of Ser5 results in transcription initiation and phosphorylation of Ser2 leads to elongation. We wanted to determine if PFI-90 treatment and the resulting increase in H3K9me2 was associated with disruption of RNA Pol2. We performed ChIP-seq analysis and found that PFI-90 treatment resulted in remarkable decrease in RNA Pol2 binding at TSS and phosphorylation of RNA Pol2 at both Ser5 and Ser2 (Fig. 6A). Furthermore, we saw decrease in enhancer bound RNA Pol2 and phosphorylation (Fig. 6B). Looking at MYOD1, there was significant decrease in RNA Pol2 binding and phosphorylation both at gene body and enhancers which was associated with increased H3K9me2 (Fig. 6C). Next, we determined the differential RNA Pol2 peaks between PFI-90 and DMSO and ranked the associated genes. GSEA analysis showed that PAX3-FOXO1 gene sets were enriched in downregulated peaks for Total Pol2 and Pol2 Ser5 consistent with RNA-seq findings of downregulated PAX3-FOXO1 target transcripts (Fig. 6D and Supplementary Table 5, Table 4–5).
PFI-90 Inhibits FP-RMS Tumor Growth In Vivo
Finally, we tested PFI-90 in two xenograft models of FP-RMS using the RH4 cell line. In a metastatic intravenous mouse model, PFI-90 was able to significantly delay tumor progression measured by luciferase signal (Fig. 7A, p = 0.0016). We also tested PFI-90 in an orthotopic model of FP-RMS injecting tumor cells intramuscularly into the gastrocnemius muscle. We again saw a significant delay in tumor progression measured by tumor volume (Fig. 7B, p = 0.0046). There was no significant change in weight during PFI-90 treatment (Supplementary Fig. 11). We also performed an additional in vivo study using a subcutaneous tumor model. After 5 days of daily treatment with PFI-90, tumors were collected for RNA-seq analysis. GSEA showed downregulation of PAX3-FOXO1 nuclear targets, and enrichment for myogenesis and apoptosis gene sets after PFI-90 treatment versus DMSO, consistent with our in vitro findings (Fig. 7C and Supplementary Table 6).