BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models

Anastasia Wyce, Gopinath Ganji, Kimberly N Smitheman, Chun-Wa Chung, Susan Korenchuk, Yuchen Bai, Olena Barbash, BaoChau Le, Peter D Craggs, Michael T McCabe, Karen M Kennedy-Wilson, Lydia V Sanchez, Romain L Gosmini, Nigel Parr, Charles F McHugh, Dashyant Dhanak, Rab K Prinjha, Kurt R Auger, Peter J Tummino, Anastasia Wyce, Gopinath Ganji, Kimberly N Smitheman, Chun-Wa Chung, Susan Korenchuk, Yuchen Bai, Olena Barbash, BaoChau Le, Peter D Craggs, Michael T McCabe, Karen M Kennedy-Wilson, Lydia V Sanchez, Romain L Gosmini, Nigel Parr, Charles F McHugh, Dashyant Dhanak, Rab K Prinjha, Kurt R Auger, Peter J Tummino

Abstract

BET family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective inhibitors of BET proteins exhibit potent anti-proliferative activity in a number of hematologic cancer models, in part through suppression of the MYC oncogene and downstream Myc-driven pathways. However, little is currently known about the activity of BET inhibitors in solid tumor models, and whether down-regulation of MYC family genes contributes to sensitivity. Here we provide evidence for potent BET inhibitor activity in neuroblastoma, a pediatric solid tumor associated with a high frequency of MYCN amplifications. We treated a panel of neuroblastoma cell lines with a novel small molecule inhibitor of BET proteins, GSK1324726A (I-BET726), and observed potent growth inhibition and cytotoxicity in most cell lines irrespective of MYCN copy number or expression level. Gene expression analyses in neuroblastoma cell lines suggest a role of BET inhibition in apoptosis, signaling, and N-Myc-driven pathways, including the direct suppression of BCL2 and MYCN. Reversal of MYCN or BCL2 suppression reduces the potency of I-BET726-induced cytotoxicity in a cell line-specific manner; however, neither factor fully accounts for I-BET726 sensitivity. Oral administration of I-BET726 to mouse xenograft models of human neuroblastoma results in tumor growth inhibition and down-regulation MYCN and BCL2 expression, suggesting a potential role for these genes in tumor growth. Taken together, our data highlight the potential of BET inhibitors as novel therapeutics for neuroblastoma, and suggest that sensitivity is driven by pleiotropic effects on cell growth and apoptotic pathways in a context-specific manner.

Conflict of interest statement

Competing Interests: The authors have read the journal's competing interest policy and have the following conflicts to disclose: all authors are current or former employees of GlaxoSmithKline. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. I-BET726: a novel selective inhibitor…
Figure 1. I-BET726: a novel selective inhibitor of BET family proteins.
(a) Chemical structure of GSK1324726A (I-BET726). (b) Crystal structure of I-BET726 (magenta) bound to the acetyl-binding pocket of BRD4-BD1 (resolution: 1.6 Å). (c) Concentration response curves for determination of binding affinity of I-BET726 to BRD2, BRD3, and BRD4 bromodomains by ligand displacement detected using Time Resolved Fluorescence Resonance Energy Transfer (TR-FRET). IC50 values for BRD2, BRD3, and BRD4 are indicated. (d) Selectivity profile of I-BET726 showing average temperature shifts (delta Tm) in degrees Celsius for a panel of bromodomain proteins using a fluorescent thermal shift assay. N= 2 for all proteins except CREBBP (n= 4).
Figure 2. I-BET726 treatment results in potent…
Figure 2. I-BET726 treatment results in potent growth inhibition and cytotoxicity in neuroblastoma cell lines.
(a) gIC50 values observed for I-BET726 in a panel of neuroblastoma cell lines obtained from a 6 day growth-death assay. (b) Concentration response curves for SK–N-AS and CHP-212 from 6 day growth-death assay. Black horizontal line indicates growth in DMSO-treated controls. Red line indicates T0 value (100%). gIC50 and Ymin-T0 values are indicated. Data presented as the average of two independent curves from a single experiment, and is representative of data from three independent biological replicates. (c) Ymin-T0 values observed for I-BET726 in the panel of neuroblastoma cell lines obtained from a 6 day growth-death assay. Gray bars indicate a cytotoxic response, defined by a Ymin-T0 value ≤ -50, with evidence of net cell death (Y < T0) along the growth curve at concentrations less than 6 µM. (d) Graph summarizing number of cells in G1 phase (as a percent of total cell population) in the indicated neuroblastoma cell lines following treatment with a titration of I-BET726 (5 nM-20 000 nM) for 2 days based on propidium iodide staining. V represents vehicle (DMSO) control sample. (e) Histograms generated from cell cycle analysis in the CHP-212 cell line following 4 days treatment with the indicated concentration of I-BET726. Percentage of cells in G1 phase and sub-G1 phase are indicated. (f) Caspase induction in the indicated neuroblastoma cell lines following treatment with a titration of I-BET726 for one, two, or three days. Data is presented as fold induction over DMSO controls, following normalization to total cell number as measured by CellTiter-Glo.
Figure 3. Global transcript profiling in neuroblastoma…
Figure 3. Global transcript profiling in neuroblastoma cell lines treated with I-BET726 reveals gene expression changes in apoptotic and signaling pathways.
(a) Hierarchical clustering of statistically significant probes that were differentially expressed in 100 nM or 1 µM treatments of I-BET726 relative to vehicle in CHP-212 and SK–N–SH. (b) Venn analysis for up-regulated and down-regulated probes described in (a) in the CHP-212 cell line. (c) Venn analysis for overlap of up-regulated and down-regulated probes in the SK–N–SH and CHP-212 cell lines. (d) Functional analyses of expression changes were performed by GO and canonical pathway enrichment at the gene level. A subset of statistically significant categories for GO Biological Process (>100 genes) and canonical pathways (>20 genes) from KEGG and BioCarta that were common among the two cell lines are shown. (e) qRT-PCR confirmation of a subset of genes selected from the functional analyses described in (d). Data represent mean value ± standard deviation for three independent biological replicates. Asterisks indicate statistical significance as measured by t-test (p <0.05).
Figure 4. Global transcript profiling reveals gene…
Figure 4. Global transcript profiling reveals gene expression changes in MYC-family pathways.
(a) qRT-PCR confirmation of changes in MYCN and MYC expression following treatment with the indicated concentration of I-BET726. Data represent mean value ± standard deviation for three independent biological replicates. Asterisks indicate statistical significance as measured by t-test (p <0.05). (b) GSEA enrichment plots showing the down-regulation of gene sets associated with Myc/Max and N-Myc binding motifs in I-BET726-treated CHP-212 cells. Normalized enrichment scores (NES) and FDR q values are indicated. (c) A MYCN transcriptional regulation network was constructed (see Materials and Methods) to depict N-Myc pathway genes that were modulated by I-BET726 treatment. Red and blue circles represent increased and decreased expression changes, respectively. Green, red and grey edges are shown for activation, inhibition and unspecified interaction types, respectively. (d) qRT-PCR confirmation of a subset of genes selected from the N-Myc network analysis described in (c). Data presented as described in (a).
Figure 5. MYCN expression is directly regulated…
Figure 5. MYCN expression is directly regulated by BRD4 and repressed by treatment with I-BET726.
(a) Left: Concentration response curve for MYCN RNA expression following 24 hour treatment with I-BET726 in the CHP-212 cell line. Data was normalized to GAPDH and is presented as expression relative to DMSO-treated controls. Data presented as the average of two independent curves from a single experiment, and is representative of data from three independent biological replicates. Right: Table of IC50 and percent inhibition values for MYCN suppression following 24 hour treatment with I-BET726 in the indicated cell lines. (b) BRD4 ChIP in the non-MYCN-amplified cell line SK–N–SH. Binding of BRD4 to the MYCN promoter or to an intergenic region on Chromosome 12 following treatment with vehicle or 1µM I-BET726 for six hours. Data is presented as fold enrichment over signal generated from IgG control immunoprecipitations. Data shown is from a single experiment representative of typical results. (c) BRD4 ChIP data at the MYCN promoter, presented as percent of vehicle control signal in the non-MYCN-amplified cell line SK–N–SH (left) and the MYCN-amplified cell line CHP-212 (right). SK–N–SH data represents the mean value ± standard deviation for three independent biological replicates. Asterisk indicates statistical significance as measured by t-test (p= 0.005). CHP-212 data represents the mean value ± standard deviation for two independent biological replicates. (d) Western blot analysis of N-Myc expression in the MYCN-amplified cell lines CHP-212 and IMR32 following 24 or 48 hour treatment with vehicle or 1µM I-BET726. Actin expression included as a loading control. (e) gIC50 values obtained from CHP-212 cells overexpressing GFP or MYCN following treatment with I-BET726 in a 6 day growth-death assay. Data represents the mean value ± standard deviation from four independent experiments. (f) Concentration response curves for GFP or MYCN–overexpressing CHP-212 cells from a 6 day growth-death assay. Horizontal line indicates T0 measurement (normalized to 100%). Data shown was from a single experiment representative of typical results. (g) Left: Histograms generated from cell cycle analysis in GFP- or MYCN-overexpressing CHP-212 cells following 4 days treatment with 5 µM I-BET726. Right: Percentage of cells in subs G1, G1, S, and G2 phases from the cell cycle experiment.
Figure 6. Suppression of BCL2 expression by…
Figure 6. Suppression of BCL2 expression by I-BET726.
(a) Left: Concentration response curve for BCL2 RNA expression following 24 hour treatment with I-BET726 in the CHP-212 cell line. Data was normalized to GAPDH and presented as expression relative to DMSO-treated controls. Data presented as the average of two independent curves from a single experiment, and is representative of data from two independent biological replicates. Right: Table of IC50 values and percent inhibition of BCL2 expression following 24 hour treatment with I-BET726. (b) BRD4 ChIP in the non-amplified neuroblastoma cell line SK–N–SH. Binding of BRD4 to the BCL2 promoter or to an intergenic region on Chromosome 12 following treatment with vehicle or 1 µM I-BET726 for six hours. Data is presented as fold enrichment over signal generated from IgG control immunoprecipitations. Data shown was from a single experiment representative of typical results. (c) BRD4 ChIP data at the BCL2 promoter, presented as percent of vehicle control signal. Data represent the mean value ± standard deviation for three independent biological replicates. Asterisk indicates statistical significance as measured by T-test (p= 0.002). (d) Western blot analysis of Bcl-2 expression in the MYCN-amplified cell lines CHP-212 and IMR32 following 48 hour treatment with vehicle or 1 µM I-BET726. Tubulin expression included as a loading control. (e) gIC50 values obtained from CHP-212 or LA-N-2 cells overexpressing GFP or BCL2 following treatment with I-BET726 in a 6 day growth-death assay. Data represents the mean value ± standard deviation from three independent experiments. (f) Ymin-T0 values for CHP-212 or LA-N-2 cells overexpressing GFP or BCL2. Data represents the mean value ± standard deviation from three independent experiments. Asterisk indicates statistical significance as measured by t-test (p= 0.02). (g) Western blot analysis of Bcl-2 expression in CHP-212 or LA-N-2 cells overexpressing GFP or BCL2 following 48 hour treatment with DMSO or 1 µM I-BET726. Actin expression included as a loading control.
Figure 7. Analysis of I-BET726 activity in…
Figure 7. Analysis of I-BET726 activity in vivo.
(a) Mean absolute body weight ± SD for mice in the SK–N-AS (left) and CHP-212 (right) xenograft studies treated with vehicle, 5 mg/kg, or 15 mg/kg I-BET726. (b) Mean absolute tumor volumes ± SEM for SK–N-AS subcutaneous xenografts following treatment with 5 mg/kg or 15 mg/kg I-BET726. Asterisks indicate statistical significance as measured by t-test (p <0.05). Tumor growth inhibition (TGI) for the 15 mg/kg group was 58% on day 14 (n= 9; p= 0.0060). (c) Mean absolute tumor volumes ± SEM for CHP-212 subcutaneous xenografts following treatment with 5 mg/kg or 15 mg/kg I-BET726. Asterisks indicate statistical significance as measured by T-test (p <0.05). TGI for 5 mg/kg was 50% on Day 42 (n= 8; p= 0.1816). TGI for 15 mg/kg was 82% on Day 42 (n=5; p =0.0488). (d) Pharmacodynamic analysis in CHP-212 and SK–N-AS xenografts 8 hours after initial dose of I-BET726. qRT-PCR analysis of MYCN, MYC, and BCL2 expression following I-BET726 treatment in the indicated models. Data is presented as fold induction compared to vehicle treated controls, and represents the average ± SD of data from three animals. (e) qRT-PCR analysis of apoptotic pathway and N-Myc pathway genes in CHP-212 xenografts 8 hours following treatment with I-BET726 on day 8 of study. Data is presented as described in (d).

References

    1. Rodríguez-Paredes M, Esteller M (2011) Cancer epigenetics reaches mainstream oncology. Nat Med 17: 330-339. PubMed: .
    1. Jenuwein T, Allis CD (2001) Translating the histone code. Science 293: 1074-1080. doi:. PubMed: .
    1. Florence B, Faller DV (2001) You bet-cha: a novel family of transcriptional regulators. Front Biosci 6: D1008-D1018. doi:. PubMed: .
    1. Dey A, Nishiyama A, Karpova T, McNally J, Ozato K (2009) Brd4 marks select genes on mitotic chromatin and directs postmitotic transcription. Mol Biol Cell 20: 4899-4909. doi:. PubMed: .
    1. Yang Z, He N, Zhou Q (2008) Brd4 recruits P-TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol Cell Biol 28: 967-976. doi:. PubMed: .
    1. Denis GV, Vaziri C, Guo N, Faller DV (2000) RING3 kinase transactivates promoters of cell cycle regulatory genes through E2F. Cell Growth Differ 11: 417-424. PubMed: .
    1. Denis GV, McComb ME, Faller DV, Sinha A, Romesser PB et al. (2006) Identification of transcription complexes that contain the double bromodomain protein Brd2 and chromatin remodeling machines. J Proteome Res 5: 502-511. doi:. PubMed: .
    1. Jones MH, Numata M, Shimane M (1997) Identification and characterization of BRDT: A testis-specific gene related to the bromodomain genes RING3 and Drosophila fsh. Genomics 45: 529-534. doi:. PubMed: .
    1. LeRoy G, Rickards B, Flint SJ (2008) The double bromodomain proteins Brd2 and Brd3 couple histone acetylation to transcription. Mol Cell 30: 51-60. doi:. PubMed: .
    1. Chung CW, Coste H, White JH, Mirguet O, Wilde J et al. (2011) Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J Med Chem 54: 3827-3838. doi:. PubMed: .
    1. Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M et al. (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478: 529-533. doi:. PubMed: .
    1. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB et al. (2010) Selective inhibition of BET bromodomains. Nature 468: 1067-1073. doi:. PubMed: .
    1. Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S et al. (2010) Suppression of inflammation by a synthetic histone mimic. Nature 468: 1119-1123. doi:. PubMed: .
    1. Prinjha RK, Witherington J, Lee K (2012) Place your BETs: the therapeutic potential of bromodomains. Trends Pharmacol Sci 33: 146-153. doi:. PubMed: .
    1. French CA (2010) NUT midline carcinoma. Cancer Genet Cytogenet 203: 16-20. doi:. PubMed: .
    1. Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H et al. (2011) RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478: 524-528. doi:. PubMed: .
    1. Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S et al. (2011) Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A 108: 16669-16674. doi:. PubMed: .
    1. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J et al. (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146: 904-917. doi:. PubMed: .
    1. Ott CJ, Kopp N, Bird L, Paranal RM, Qi J et al. (2012) BET bromodomain inhibition targets both c-MYC and IL7R in high-risk acute lymphoblastic leukemia. Blood, 120: 2843–52. PubMed: .
    1. Albihn A, Johnsen JI, Henriksson MA (2010) MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res 107: 163-224. doi:. PubMed: .
    1. Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S et al. (2010) The landscape of somatic copy-number alteration across human cancers. Nature 463: 899-905. doi:. PubMed: .
    1. Vita M, Henriksson M (2006) The Myc oncoprotein as a therapeutic target for human cancer. Semin Cancer Biol 16: 318-330. doi:. PubMed: .
    1. Darnell JE Jr. (2002) Transcription factors as targets for cancer therapy. Nat Rev Cancer 2: 740-749. doi:. PubMed: .
    1. Chen QR, Song YK, Yu LR, Wei JS, Chung JY et al. (2010) Global genomic and proteomic analysis identifies biological pathways related to high-risk neuroblastoma. J Proteome Res 9: 373-382. doi:. PubMed: .
    1. Fredlund E, Ringnér M, Maris JM, Påhlman S (2008) High Myc pathway activity and low stage of neuronal differentiation associate with poor outcome in neuroblastoma. Proc Natl Acad Sci U S A 105: 14094-14099. doi:. PubMed: .
    1. Raetz EA, Kim MK, Moos P, Carlson M, Bruggers C et al. (2003) Identification of genes that are regulated transcriptionally by Myc in childhood tumors. Cancer 98: 841-853. doi:. PubMed: .
    1. Wang Y, Sugiyama H, Axelson H, Panda CK, Babonits M et al. (1992) Functional homology between N-myc and c-myc in murine plasmacytomagenesis: plasmacytoma development in N-myc transgenic mice. Oncogene 7: 1241-1247. PubMed: .
    1. Malynn BA, de Alboran IM, O’Hagan RC, Bronson R, Davidson L et al. (2000) N-myc can functionally replace c-myc in murine development, cellular growth, and differentiation. Genes Dev 14: 1390-1399. PubMed: .
    1. Zeller KI, Jegga AG, Aronow BJ, O’Donnell KA, Dang CV (2003) An integrated database of genes responsive to the Myc oncogenic transcription factor: identification of direct genomic targets. Genome Biol 4: R69. doi:. PubMed: .
    1. Reed JC, Meister L, Tanaka S, Cuddy M, Yum S et al. (1991) Differential expression of bcl2 protooncogene in neuroblastoma and other human tumor cell lines of neural origin. Cancer Res 51: 6529-6538. PubMed: .
    1. Castle VP, Heidelberger KP, Bromberg J, Ou X, Dole M et al. (1993) Expression of the apoptosis-suppressing protein bcl-2, in neuroblastoma is associated with unfavorable histology and N-myc amplification. Am J Pathol 143: 1543-1550. PubMed: .
    1. Maris JM, Hogarty MD, Bagatell R, Cohn SL (2007) Neuroblastoma. Lancet 369: 2106-2120. doi:. PubMed: .
    1. Lockwood WW, Zejnullahu K, Bradner JE, Varmus H (2012) Sensitivity of human lung adenocarcinoma cell lines to targeted inhibition of BET epigenetic signaling proteins. Proc Natl Acad Sci U S A 109: 19408-19413. doi:. PubMed: .
    1. Cheng Z, Gong Y, Ma Y, Lu K, Lu X et al. (2013) Inhibition of BET Bromodomain Targets Genetically Diverse Glioblastoma. Clin Cancer Res, 19: 1748–59. PubMed: .
    1. Jiang R, Xue S, Jin Z (2011) Stable knockdown of MYCN by lentivirus-based RNAi inhibits human neuroblastoma cells growth in vitro and in vivo. Biochem Biophys Res Commun 410: 364-370. doi:. PubMed: .
    1. Galderisi U, Di BG, Cipollaro M, Peluso G, Cascino A et al. (1999) Differentiation and apoptosis of neuroblastoma cells: role of N-myc gene product. J Cell Biochem 73: 97-105. doi:. PubMed: .
    1. Strieder V, Lutz W (2003) E2F proteins regulate MYCN expression in neuroblastomas. J Biol Chem 278: 2983-2989. doi:. PubMed: .
    1. Valentijn LJ, Koster J, Haneveld F, Aissa RA, van Sluis P et al. (2012) Functional MYCN signature predicts outcome of neuroblastoma irrespective of MYCN amplification. Proc Natl Acad Sci U S A 109: 19190-19195. doi:. PubMed: .
    1. Godfried MB, Veenstra M, Sluis P, Boon K, Asperen R et al. (2002) The N-myc and c-myc downstream pathways include the chromosome 17q genes nm23-H1 and nm23-H2. Oncogene 21: 2097-2101. doi:. PubMed: .
    1. Puissant A, Frumm SM, Alexe G, Bassil CF, Qi J et al. (2013) Targeting MYCN in Neuroblastoma by BET Bromodomain Inhibition. Cancer Discov.
    1. Lamers F, Schild L, den Hartog IJ, Ebus ME, Westerhout EM et al. (2012) Targeted BCL2 inhibition effectively inhibits neuroblastoma tumour growth. Eur J Cancer, 48: 3093–103. PubMed: .
    1. Dole M, Nuñez G, Merchant AK, Maybaum J, Rode CK et al. (1994) Bcl-2 inhibits chemotherapy-induced apoptosis in neuroblastoma. Cancer Res 54: 3253-3259. PubMed: .
    1. van Golen CM, Castle VP, Feldman EL (2000) IGF-I receptor activation and BCL-2 overexpression prevent early apoptotic events in human neuroblastoma. Cell Death Differ 7: 654-665. doi:. PubMed: .
    1. Lestini BJ, Goldsmith KC, Fluchel MN, Liu X, Chen NL et al. (2009) Mcl1 downregulation sensitizes neuroblastoma to cytotoxic chemotherapy and small molecule Bcl2-family antagonists. Cancer Biol Ther 8: 1587-1595. doi:. PubMed: .
    1. Jasty R, van Golen C, Lin HJ, Solomon G, Heidelberger K et al. (2001) Bcl-2 and M-Myc coexpression increases IGF-IR and features of malignant growth in neuroblastoma cell lines. Neoplasia 3: 304-313. doi:. PubMed: .
    1. Brambilla E, Negoescu A, Gazzeri S, Lantuejoul S, Moro D et al. (1996) Apoptosis-related factors p53, Bcl2, and Bax in neuroendocrine lung tumors. Am J Pathol 149: 1941-1952. PubMed: .
    1. McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C et al. (2012) EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492: 108-112. doi:. PubMed: .
    1. Vindeløv LL, Christensen IJ, Nissen NI (1983) A detergent-trypsin method for the preparation of nuclei for flow cytometric DNA analysis. Cytometry 3: 323-327. doi:. PubMed: .
    1. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P et al. (2006) GenePattern. Nat Genet 2 0: 38: 500-501
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL et al. (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102: 15545-15550. doi:. PubMed: .
    1. Clements EG, Mohammad HP, Leadem BR, Easwaran H, Cai Y et al. (2012) DNMT1 modulates gene expression without its catalytic activity partially through its interactions with histone-modifying enzymes. Nucleic Acids Res, 40: 4334–46. PubMed: .

Source: PubMed

3
Prenumerera