The quassinoid analogue NBT-272 has been reported to inhibit MYC, thus warranting a further effort 7to better understand its preclinical properties in models of embryonal tumors (ET), a family of childhood malignancies sharing relevant biological and genetic features such as deregulated expression of MYC oncogenes. In our study, NBT-272 displayed a strong antiproliferative activity in vitro that resulted from the combination of diverse biological effects, ranging from G1/S arrest of the cell cycle to apoptosis and autophagy. The compound prevented the full activation of both eukaryotic translation initiation factor 4E (eIF4E) and its binding protein 4EBP-1, regulating cap-dependent protein translation. Interestingly, all responses induced by NBT-272 in ET could be attributed to interference with 2 main proproliferative signaling pathways, that is, the AKT and the MEK/extracellular signal-regulated kinase pathways. These findings also suggested that the depleting effect of NBT-272 on MYC protein expression occurred via indirect mechanisms, rather than selective inhibition. Finally, the ability of NBT-272 to arrest tumor growth in a xenograft model of neuroblastoma plays a role in the strong antitumor activity of this compound, both in vitro and in vivo, with its potential to target cell-survival pathways that are relevant for the development and progression of ET. Mol Cancer Ther; 9(12); 3145–57. ©2010 AACR.

Embryonal tumors (ET) represent an important group of childhood malignancies arising from different tissues of fetal origin (1, 2). Medulloblastoma (MB) is a tumor of the central nervous system, originating from granular progenitor cells of the cerebellum (3) and accounting for about 20% of all childhood brain tumors (4). Neuroblastoma (NB) is the most common extracranial solid cancer in children originating from the sympathetic nervous system (5, 6). Constant progress has been achieved in clinical practice by increasing the overall survival rate of MB and NB patients, although novel therapeutic strategies are still needed, particularly to treat the high-risk patients characterized by a more aggressive disease and poor prognosis. In both MB and NB, high-grade malignancy is often associated with overexpression or amplification of MYC oncogenes (7–11) playing a role in embryogenesis and, if aberrantly expressed, in tumor development as well (12).

There is a long history of therapeutic benefits produced by natural compounds and some of these have formed the backbone for further targeted chemical modifications improving efficacy and selectivity. Quassinoids have been investigated for their antineoplastic properties (13), which has led to the identification of interesting candidates such as bruceantin, with significant inhibitory effects on leukemia, lymphoma, and myeloma cell growth, both in vitro and in vivo (14). Bruceantin was reported to induce apoptosis and cell differentiation, thus justifying efforts to develop quassinoid analogues with improved efficacy.

Among the derivatives of bruceantin, NBT-272 displayed a significantly stronger toxicity than the parent molecule, as tested in different models of hematological and solid tumors (15, 16). Moreover, NBT-272 was reported to induce downregulation of c-MYC in MB-derived cells (16), thus raising the question of the mechanisms underlying NBT-272's cytotoxicity and its selectivity for MYC in ET.

Besides activating programmed cell death, quassinoids were shown to arrest cell growth by impairing protein synthesis mediated by the eukaryotic translation initiation factor 4E (eIF4E; ref. 17). The phosphorylation status of eIF4E plays an important biological role in modulation of normal cell growth in response to extracellular stimuli and during development (18). However, eIF4E was also reported to be an oncogene (19) and its transformation properties were attributed, at least in part, to induction of cell cycle progression through cyclin D1 (CCND1) and MYC (20). Activation of eIF4E itself is mainly regulated by 2 pathways, namely the mitogen-activated protein kinase pathway (21) and the AKT/mammalian target of rapamycin (mTOR) pathway via the 4E binding protein (4EBP-1; ref. 22).

In this study, we have investigated the effect of NBT-272 in different cellular models of ET, both in vitro and in vivo. The compound triggered multiple cellular responses, ranging from cell cycle arrest to induction of autophagy, which are downstream effector mechanisms of the AKT/mTOR and extracellular signal-regulated kinase (ERK) pathways. In turn, interference with these signaling events could also explain the inhibitory effect of NBT-272 on protein synthesis and on expression of c-MYC/MYCN in different ET entities.

Cell lines

The following human cell lines were used: the NB SK-N-BE, SH-SY5Y, and WAC-2 (23; G. M. Brodeur, Children's Hospital of Philadelphia, Philadelphia, PA); retinoblastoma (RB) Y79 and WERI (A. Eggert, University Children's Hospital, Essen, Germany); Ewing's sarcoma family of tumors (ESFT) A673 and TC71 (H. Kovar, St. Anna Children's Cancer Research Institute, Vienna, Austria); and malignant rhabdoid tumor (MRT) LP and MON (O. Delattre, Institut Curie, Paris, France). MB DAOY and D341 were purchased from American Type Culture Collection. Cells that were not purchased from the American Type Culture Collection were authenticated by comparative genomic hybridization (in the laboratories mentioned above or as described below). The clone DAOY M2 (overexpressing c-MYC) was kindly provided by D. Stearns (Johns Hopkins University, Baltimore, MD; ref. 11). NB, ESFT, and MRT cell lines were grown in RPMI medium (Life Technologies/Invitrogen) supplemented with 10% (v/v) fetal calf serum (FCS) and penicillin/streptomycin (PS)/l-glutamine, whereas RB cells were grown in Dulbecco's modified Eagle's medium (DMEM; Invitrogen) supplemented with 15% (v/v) FCS Gold (Invitrogen), 10 μg/mL insulin (Sigma-Aldrich), 50 μmol/L 2-β-mercaptoethanol (Sigma-Aldrich), and PS/l-glutamine. MB cells were cultured in Richter's zinc option medium (Invitrogen) supplemented with 10% (v/v) FCS and PS/l-glutamine.

Array CGH

The ET-derived cell lines used in the study were profiled by array comparative genomic hybridization (aCGH) using a custom 60K Embryonal Tumor array CGH-chip (ET-aCGH chip; Agilent Technologies), as described by Kumps et al. (24).

Cell viability, clonogenic, and apoptosis assays

The 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS)-based CellTiter 96 AQueous One assay (Promega) was used to quantify cell viability. Colony formation in agar was tested by seeding single cells (4,000 per well) in agar-containing medium [0.35% (w/v) final concentration] on top of a 0.5% (w/v) agar layer. Apoptosis was investigated by quantifying the activation of caspases, by using the Caspase-Glo 3/7 Assay (Promega) according to manufacturer's instructions. The In Situ Cell Death Detection Kit (Roche) was also employed to evaluate the proapoptotic response of ET cells. The terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling (TUNEL) reaction was done in the presence of fluorescein-labeled nucleotides and followed by detection under a fluorescent microscope (Axioskop 2; Zeiss).

Cell cycle analysis

Cells [(5–10) × 105] were treated for the indicated time intervals with different doses of NBT-272 or were mock treated (control). After washing with PBS, the cells were washed and harvested in a 2 mmol/L EDTA/PBS solution. Cell pellets were fixed with ice-cold 70% ethanol on ice for 40 to 60 minutes and, after centrifugation at 2,500 g for 5 minutes, resuspended with 50 μg/mL propidium iodide/PBS containing 100 U/mL RNAase (Sigma-Aldrich). The DNA content was estimated by using a BD FACS Canto II flow cytometry system for acquisition and the FlowJo software for the data analysis.

Gene expression analysis by PCR arrays

Total RNA from DAOY, D341, SK-N-BE, and SH-SY-5Y cells, treated with 20 nmol/L NBT-272 for 24 hours or left untreated, was subjected to reverse transcriptase (RT; RT2 First Strand Kit; SABiosciences). The resulting cDNA was used as a template for RT2 Profiler PCR Array (SABiosciences) for pathway-focused gene expression analysis (using sets of arrayed TaqMan probes for cell cycle and apoptosis genes). Gene expression was expressed as log2 ratio (where ratio is 2−ΔΔCt), relative to normalization to untreated control samples and to the average expression of five housekeeping genes (B2M, HPRT1, RPL13A, GAPDH, and ACTB). Only statistically significant gene expression changes over 3 independent experiments were considered.

Western blot analysis

Total cell lysates were prepared in a radioimmunoprecipitation assay buffer buffer [50 mmol/L Tris-HCl (pH 7.5), 0.1% sodium laurilsulfate, 0.1% sodium deoxycholate, 150 mmol/L NaCl, 1% NP40, 1 mmol/L EDTA, and 1 mmol/L EGTA], supplemented with protease inhibitors (Roche) and phosphatase inhibitors (10 mmol/L sodium fluoride, 10 mmol/L sodium orthovanadate, and 10 mmol/L sodium β-glycerophosphate). The amount of protein was determined by BCA Protein Assay (Pierce). Cell lysates in SDS-sample buffer were boiled for 8 minutes and equal protein amounts were resolved by 4% to 12% gradient SDS-PAGE and immunoblotting using polyvinylidene difluoride membranes. The antibodies reacting against the following proteins were employed: CCND1, caspase-3, AKT, mTOR, phospho-ERK 1/2(Tyr202/204; Santa Cruz Biotechnology); ERK, phospho-AKT (Ser473), phospho-MEK1/2 (Ser217/221), phospho-RB (Ser795), RB, phospho-S6 (Ser235/236), S6, phospho-mTOR (Ser2248), phospho-MNK1 (Thr197/202), MNK1, phospho-eIF4E (Ser209), eIF4E, 4EBP-1, phospho-4EBP-1 (Thr37/46), cyclin E, CDK2, CDK4, PARP, LC-3, c-MYC, MYCN, and ATG5 (Cell Signaling); β-tubulin and β-actin antibodies were purchased from Sigma-Aldrich, whereas the anti-SQSTM1/p62 was purchased from BIOMOL International. To inhibit proteasome-dependent protein degradation, cells were treated with 50 μmol/L MG-132 (Sigma-Aldrich). The PI3K/mTOR inhibitor BEZ-235 was purchased by Axon Medchem.

Transmission electron microscopy

After treatment (with NBT-272, rapamycin, or vehicle) for different time intervals, DAOY and SK-N-BE cells were subjected to fixation at room temperature for 30 minutes with 2% glutaraldehyde and 0.8% formaldehyde in 50 mmol/L sodium cacodylate (pH 7.3), followed by 30-minute exposure to 1% OsO4 and 1.5% K4Fe(CN)6 in 50 mmol/L sodium cacodylate (pH 7.3). Fixed samples were contrasted using 2% uranyl acetate in water for 2 hours, progressively dehydrated in increasing concentrations of ethanol, and embedded into epon (Catalys). Ultrathin sections of 50 nm were prepared and contrasted with uranyl acetate and lead citrate. Samples were analyzed by using a CM 100 transmission electron microscope (TEM; Philips).

In vivo xenograft model

Human NB SH-SY5Y cells were stably transfected with a pLentiV5-Luciferase-expressing vector (Invitrogen) as described elsewhere (25). SH-SY5Y-LUC clones were grown under standard conditions in DMEM containing 10% (v/v) FCS, l-glutamine, and blasticidine for selection.

Eight-week-old female athymic nude mice from Harlan Laboratories were inoculated subcutaneously in the right rear flank with 2 × 106 SHSY5Y-LUC cells, injected in 100 μL PBS solution. Tumor cell growth was monitored weekly by measuring luminescence emission, referred to as bioluminescence imaging (BLI) levels, using the IVIS 3D Illumina Imaging System (Xenogen Corp.). A detailed procedure can be obtained from Caliper. The BLI/tumor size ratio was quantified by encircling the luciferase-emitting areas using the Living Images Software 3.2 (Xenogen). Three weeks after implantation, the mice were divided into 2 homogeneous groups based on their BLI level (photon/s/cm2). Each group received either 10 μmol/L per week of NBT-272 or vehicle (EtOH 0.003%/PBS), administered as 100 μL solution each time, 3 times a week. The analysis of the tumor growth was done 3, 4, and 6 weeks after the first NBT-272 administration. The dose was adjusted by taking into account preliminary data provided by NaPro BioTherapeutics, Inc. (maximum tolerated dose = 0.9 mg/kg/wk; ref. 15) and the in vitro results. The treatment was done for 6 weeks before the animals were sacrificed, and tumor samples were analyzed by immunohistochemistry and Western blotting. Quantitative data analysis of the tumor size was done by ANOVA statistical tests, using the Statview program.

All animal experiments were conducted according to national and international guidelines and following approval by the Institutional Animal Care and Ethical Committee of CEINGE-University of Naples Federico II (Protocol 29, September 30, 2009) and of the Italian Ministry of Health (Dipartimento Sanità Pubblica Veterinaria D.L. 116/92).

Immunohistochemistry

Paraffin-embedded tumor samples were treated with xylene, heated for 45 minutes (in citrate buffer, pH 6.0), and finally treated for 15 minutes with 1% H2O2 (Carlo Erba Reagents). Tissues for immunohistochemistry were blocked with 3% bovine serum albumin, 5% total goat serum, and 0.3% Tween 20/PBS (1 hour, room temperature). Primary antibodies reactive against human Ki67 (M7240; Dako, Glostrup, Denmark) and human cleaved caspase-3 (9661; Cell Signaling) were used (1 hour, room temperature, diluted 1:100 in blocking solution). After incubation with a biotin-conjugated secondary antibody (1 hour, room temperature), the detection was done in the presence of 3,3′-diaminobenzidin (Dako) according to the manufacturers' protocol. The sections were counterstained by hematoxylin/eosin (Bio Optica), washing 3 times with 0.2% Triton X-100/PBS after every incubation steps. As negative control, blocking solution was used instead of the primary antibody.

NBT-272 strongly impaired cell growth in ET-derived cell lines

NBT-272 (NaPro BioTherapeutics, Inc.; Fig. 1A) is a semisynthetic analogue of bruceantin (14, 17), with 10-fold higher toxicity than the parent compound in vitro (15). Previously published data showed the ability of NBT-272 to drastically reduce the protein expression level of the oncogene c-MYC in MB-derived cell lines (16), although the exact mechanism of action remained unexplored. In this study, the biological responses induced by NBT-272 were investigated in a panel of 13 cell lines, representative of 6 different ET entities (Fig. 1). Expression of c-MYC and MYCN was evaluated in each cell line both at protein (Supplementary Fig. S1A) and mRNA levels (Supplementary Fig. S1B); these varied significantly, reflecting the gene expression ranging from single copy to gene amplification.

Figure 1.

Dose-dependent effect of NBT-272 on cell viability and colony formation in ET-derived cell lines. A, chemical structure of NBT-272. B, MTS assays were done 72 hours after addition of increasing concentrations of NBT-272 (1–182 nmol/L) in 13 ET-derived cell lines. C, five cell lines were evaluated for anchorage-independent growth in soft agar. NBT-272 reduced colony formation by 50% relative to monolayer cultures at concentrations lower than/similar to IC50 values. IC50 (nmol/L): WAC-2, 2.3; SK-N-BE, 4.3; A673, 4.6; MON, < 1; Wit-49, 9.7.

Figure 1.

Dose-dependent effect of NBT-272 on cell viability and colony formation in ET-derived cell lines. A, chemical structure of NBT-272. B, MTS assays were done 72 hours after addition of increasing concentrations of NBT-272 (1–182 nmol/L) in 13 ET-derived cell lines. C, five cell lines were evaluated for anchorage-independent growth in soft agar. NBT-272 reduced colony formation by 50% relative to monolayer cultures at concentrations lower than/similar to IC50 values. IC50 (nmol/L): WAC-2, 2.3; SK-N-BE, 4.3; A673, 4.6; MON, < 1; Wit-49, 9.7.

Close modal

The ET cell lines were profiled for copy number changes (gain/loss) using aCGH. In Supplementary Fig. S2, the aCGH data of 8 cell lines are reported. Some cell lines displayed very few genomic aberrations (1q gain in the MRT cells LP, and 1q gain and 8q gain in MB D341), whereas others have very complex profiles, such as WERI cells with a 1q gain region that appeared very discontinuous.

The effect of NBT-272 was first evaluated in terms of cell viability, clearly showing a very potent effect of the compound in all selected ET-derived cell lines (Fig. 1B; Table 1). The IC50 values, calculated from the dose–response curves, varied in the nmol/L range of concentration, that is, about 2 orders of magnitude lower compared with many small-molecule inhibitors investigated as anticancer drugs. Additionally, 5 representative ET cell lines (WAC-2, SK-N-BE, A673, MON, Wit-49) were tested for anchorage-independent growth in soft agar in the presence of NBT-272 (Fig. 1C). The compound reduced colony formation by 50% (compared with mock-treated controls) at doses that were similar or lower than the IC50 obtained in cell viability assays (Table 1).

Table 1.

The IC50 values relative to NBT-272 toxicity, as calculated from the curves fitting the data in Figure 1B

ET entityCell line[NBT-272]IC50(nmol/L)
Medulloblastoma (MB) DAOY 13.6 
 DAOY M2 4.9 
 D341 1.5 
Neuroblastoma (NB) WAC-2 4.8 
 SK-N-BE 1.8 
 SH-SY5Y 3.5 
Retinoblastoma (RB) WERI 4.4 
 Y79 5.4 
Wilms' tumor (WT) Wit-49 4.9 
Ewing sarcoma family of tumors (ESFT) A673 6.0 
 TC71 4.6 
Malignant rhabdoid tumor (MRT) MON 11.8 
 LP 11.0 
ET entityCell line[NBT-272]IC50(nmol/L)
Medulloblastoma (MB) DAOY 13.6 
 DAOY M2 4.9 
 D341 1.5 
Neuroblastoma (NB) WAC-2 4.8 
 SK-N-BE 1.8 
 SH-SY5Y 3.5 
Retinoblastoma (RB) WERI 4.4 
 Y79 5.4 
Wilms' tumor (WT) Wit-49 4.9 
Ewing sarcoma family of tumors (ESFT) A673 6.0 
 TC71 4.6 
Malignant rhabdoid tumor (MRT) MON 11.8 
 LP 11.0 

Note: DAOY M2 cells (overexpressing c-MYC) are also reported.

NBT-272-induced cell cycle arrest at the G1/S transition

To identify which molecular characteristics were responsible for NBT-272's toxicity, we sought to investigate the diverse biological responses triggered in ET cell lines by the compound. The effect on cellular distribution at different stages of the cell cycle was evaluated by DNA staining in MB and NB cells. In dose–response experiments at different time points, 5 nmol/L NBT-272 was already sufficient to almost halve the population of replicating DAOY cells after 24 hours, with a concomitant increase in G1 population (Supplementary Fig. S3). This observation was found to extend to NB cell lines (WAC-2, SK-N-BE, and SH-SY5Y; Fig. 2A). Here, NBT-272 induced a similar effect after 16-hour treatment, thus suggesting that blocking the cell cycle progression at the G1 to S transition occurs as one of the earliest mechanisms leading to proliferative arrest.

Figure 2.

Induction of cell cycle arrest in MB and NB cells. A, WAC-2, SK-N-BE, and SH-SY5Y cells were treated with the indicated amount of NBT-272 for 16 hours or were left untreated (control). The percentage of cells in different stages of the cell cycle is reported in each diagram. B, WAC-2 and DAOY cells were subjected to treatment with 20 nmol/L NBT-272 and analyzed for expression of cell cycle–regulatory proteins by immunoblotting at different time points. The experiment was also confirmed in SH-SY5Y cells treated for 16 hours.

Figure 2.

Induction of cell cycle arrest in MB and NB cells. A, WAC-2, SK-N-BE, and SH-SY5Y cells were treated with the indicated amount of NBT-272 for 16 hours or were left untreated (control). The percentage of cells in different stages of the cell cycle is reported in each diagram. B, WAC-2 and DAOY cells were subjected to treatment with 20 nmol/L NBT-272 and analyzed for expression of cell cycle–regulatory proteins by immunoblotting at different time points. The experiment was also confirmed in SH-SY5Y cells treated for 16 hours.

Close modal

Consistent with the data obtained by cytofluorimetry, NBT-272 induced loss of CCND1 and of its interacting partner the cyclin-dependent kinase (CDK)4 (Fig. 2B). These proteins are both important regulators of the cell cycle progression through the G1 phase (26), along with the phosphorylation status of the retinoblastoma protein, which changes throughout the cell cycle with highest levels when cells enter the S phase (27). Consistent with an arrest in the G1/S transition, RB was hypophosphorylated on treatment with NBT-272 (Fig. 2B). On the contrary, the effect on CCNE1/CDK2 was transient.

Two NB cell lines (SH-SY5Y and SK-N-BE) and the MB cells DAOY and D341 were treated for 24 hours with 20 nmol/L NBT-272, followed by expression profiling of 84 cell cycle–related genes by qRT-PCR. Supplementary Fig. S4A illustrates the statistically significant gene expression changes induced by NBT-272. The 23 transcripts fell into 3 functional categories: (1) G1/S transition of the cell cycle; (2) DNA damage (e.g., growth arrest and DNA damage-inducible alpha protein, GADD45A); and (3) regulation of transcription elongation. The most relevant changes concerned genes involved in the G1 arrest, in support of the data from our biological assays (Fig. 2). For instance, consistent with a G1/S block of the cell cycle, the mRNA level of CDK2 decreased, whereas the inhibitor molecules CDKN1A/p21Cip1 and CDKN1B/p27Kip were induced on treatment (Supplementary Fig. S4A). Whereas CCND1 and CCNE1 were regulated solely at the protein level (Fig. 2B), other cyclins and CDK molecules involved in the RNA polymerase II–mediated gene transcription (CCNC/CDK8, CCNH/CDK7, and CCNT/CDK9) were found to be upregulated.

Apoptosis was induced by NBT-272 in a cell type–dependent manner

Induction of apoptosis was assessed by quantifying the activation of caspase-3/7 induced by NBT-272 (Fig. 3A). The proapoptotic response to NBT-272 varied significantly in a cell type– and tumor-dependent way. This observation was also confirmed by Cell Death ELISA assays (Roche; data not shown). Whereas NBT-272 concentrations as small as 10 nmol/L triggered apoptosis in MB-, ESFT-, and MRT-derived cell lines after 24 hours, the same doses had a very modest proapoptotic effect in other cellular models, particularly in NB (Fig. 3A). The limited proapoptotic effect of NBT-272 in MB and NB cells was also shown by immunoblotting (Fig. 3B), where caspase-3 could be only detected as full length protein and PARP cleavage was only induced in MB cell lines (e.g., in D341; Fig. 3B). This observation was confirmed by TUNEL assay, where NBT-272 was able to induce a breakage of the genomic DNA in DAOY cells (Supplementary Fig. S5).

Figure 3.

NBT-272-induced apoptosis and autophagy in ET cell lines. A, induction of apoptosis was quantified by Caspase-Glo 3/7 assay (Promega) in 10 cell lines representative of different ET entities following treatment with 10/20 nmol/L NBT-272 for 24 hours. Luminescence signals were normalized to cell viability (MTS assay). B, cleavage of PARP/caspase-3 was evaluated by Western blotting in the MB cell lines D341 and DAOY and in 2 NB cell lines (SH-SY5Y and SK-N-BE), treated as indicated. The apparent molecular weight of full length and cleaved caspase-3 on the blots is indicated (35 kDa, 17–19 kDa). C, high-resolution electron microscopy was done to analyze the effect of NBT-272 (20 nmol/L, 16 hours) on the ultrastructural organization of DAOY cells (a, c–e, and h) and of SK-N-BE cells (b, f, and g). Mock-treated cells (a, b) appeared healthy with homogeneous cytoplasm and regular nuclear envelope. Treated cells displayed a looser cytosol and membrane blebbing at the cell surface (c) and a drastic increase in the amount of vacuoles and lysosomes (c, d). Numerous autophagosomes containing cellular fragments, ribosomes, and whole mitochondria could be detected (arrow heads). DAOY cells were treated separately with rapamycin (200 nmol/L, 48 hours) as control (h). Bars, a, h = 4 μm; b–d, g = 2 μm; and e, f = 0.5 μm. D. Change in expression of the autophagy-related proteins SQSTM1 (p62) and LC-3 and of the Atg 5–12 complex (markers of autophagosome formation) was documented by immunoblotting in DAOY cells.

Figure 3.

NBT-272-induced apoptosis and autophagy in ET cell lines. A, induction of apoptosis was quantified by Caspase-Glo 3/7 assay (Promega) in 10 cell lines representative of different ET entities following treatment with 10/20 nmol/L NBT-272 for 24 hours. Luminescence signals were normalized to cell viability (MTS assay). B, cleavage of PARP/caspase-3 was evaluated by Western blotting in the MB cell lines D341 and DAOY and in 2 NB cell lines (SH-SY5Y and SK-N-BE), treated as indicated. The apparent molecular weight of full length and cleaved caspase-3 on the blots is indicated (35 kDa, 17–19 kDa). C, high-resolution electron microscopy was done to analyze the effect of NBT-272 (20 nmol/L, 16 hours) on the ultrastructural organization of DAOY cells (a, c–e, and h) and of SK-N-BE cells (b, f, and g). Mock-treated cells (a, b) appeared healthy with homogeneous cytoplasm and regular nuclear envelope. Treated cells displayed a looser cytosol and membrane blebbing at the cell surface (c) and a drastic increase in the amount of vacuoles and lysosomes (c, d). Numerous autophagosomes containing cellular fragments, ribosomes, and whole mitochondria could be detected (arrow heads). DAOY cells were treated separately with rapamycin (200 nmol/L, 48 hours) as control (h). Bars, a, h = 4 μm; b–d, g = 2 μm; and e, f = 0.5 μm. D. Change in expression of the autophagy-related proteins SQSTM1 (p62) and LC-3 and of the Atg 5–12 complex (markers of autophagosome formation) was documented by immunoblotting in DAOY cells.

Close modal

Using a similar approach to that employed for cell cycle–related genes, we investigated the transcriptional changes related to apoptosis triggered by NBT-272 (Supplementary Fig. S4B). Among the differentially expressed genes, we found key components of both the extrinsic and the intrinsic pathways of apoptosis, mainly upregulated on treatment with the compound. This was the case with proapoptotic members of the Bcl-2 family (BIK, BAD, and BAK) and with proteins bearing the caspase recruitment domain (CARD), such as Bcl-10, PYCARD and RIPK2, implicated in the recruitment and binding of caspases. Another major group, namely genes encoding for ligands and receptors of the tumor necrosis factor (TNF) superfamily, was induced (Supplementary Fig. S4B). It included TNF-α, TNF-β/LTA, TNFSF10/TRAIL, and their corresponding receptors TNFRSF10A and TNFRSF10B. Also the expression of TNFRSF5, TNFRSF9, and TNFRSF6/FAS increased on NBT-272 treatment, altogether indicating enhanced cell responsiveness to death receptor–mediated signaling.

NBT-272 triggered autophagosome formation

Because NBT-272 induced apoptosis with differing efficiency in the ET cell lines tested, we sought additional mechanisms that could lead to cell death. Formation of the distinctive traits of autophagy was investigated at the ultrastructure cellular level by TEM, thus revealing induction of a high number of double-membrane structures containing cellular material (i.e., cytosolic cargo molecules and organelles), described as autophagosomes (28).

In Fig. 3C, TEM images of DAOY (MB) and SK-N-BE (NB) cells are reproduced. Compared with mock-treated controls (Fig. 3C, a and b), treated cells (20 nmol/L NBT-272, 16 hours) displayed all the characteristic signs of autophagy induction, in particular, formation of large autophagosomes (arrow heads in Fig. 3C, e, f, and g) and increased lysosomes and vacuoles. Moreover, the cytosol appeared looser than in control cells, the nuclear envelope became irregular, the mitochondria acquired swollen crests indicative of dysfunctional activity, and myelin figures could be visualized as electron-dense structures. As positive control, DAOY cells were treated separately with rapamycin (200 nmol/L, 48 hours; Fig. 3C, h), inducing autophagy as an event associated with inhibition of mTOR (29). On comparing the effect of the 2 compounds in DAOY cells, NBT-272 proved to be a much stronger inducer of autophagy.

Formation of autophagosomes requires a rather complex rearrangement of Atg proteins, including the protein conjugation system generating the complex Atg5-Atg12 (30), which was detected by immunoblotting, on treatment of DAOY cells with 20 nmol/L NBT-272 (Fig. 3D). Concomitantly, we were able to show the degradation of sequestosome 1 (SQSTM1/p62), that is, a multifunctional adaptor molecule driving the degradation of polyubiquitinated proteins such as those tagged for autophagic clearance (31). Further, we documented the conversion of the microtubule-associated protein light chain 3 (LC3-I) into its membrane-interacting counterpart (LC3-II), which is a well-documented marker of autophagy induction.

MYC depletion resulted as an indirect effect of NBT-272

Because NBT-272 was originally described as a MYC inhibitor, we evaluated the correlation of the expression of c-MYC and MYCN in the ET cell lines (Supplementary Fig. S1) with cell sensitivity to the compound. However, only the MB cell line D341 (bearing c-MYC amplification) was significantly more sensitive to NBT-272 than DAOY cells expressing c-MYC as single copy gene (Fig. 1; ref. 16), whereas cells from other ET entities displayed a sensitivity similar to NBT-272 (Fig. 1).

Moreover, again regardless of the MYC isoform expressed and the gene copy number of c-MYC/MYCN, NBT-272 induced depletion of MYC in several ET cell lines, although exclusively at the protein level (Fig. 4A). In contrast, mRNA expression was either left almost unchanged (e.g., MYCN in SK-N-BE cells) or was enhanced (e.g., c-MYC in D341 cells) by NBT-272 in a dose-dependent manner (Fig. 4A).

Figure 4.

Indirect effect of NBT-272 on MYC. A, the effect of NBT-272 on either c-MYC or MYCN in different ET cell lines was analyzed at the protein (Western blotting) and at the mRNA level (qRT-PCR). B, cotreatment of D341 cells with MG132, inhibiting proteasome-mediated protein degradation, rescued c-MYC from the depleting effect of NBT-272. C, cell viability was evaluated in 3 NB-derived cell lines (SK-N-BE, SH-SY5Y, WAC-2) incubated for 72 hours with increasing doses of Mycro3 (34) or 10058-F4 (35). D, changes in the expression of apoptosis- (left) and cell cycle–related genes (right) were analyzed comparing the effect of NBT-272 (1) and of c-MYC downregulation by siRNA (2). For the silencing experiment, DAOY M2 cells were transfected transiently with c-MYC siRNA and gene expression was analyzed after 48 hours by using the RT2 Profiler PCR Array. The same cell line was treated for 24 hours with 20 nmol/L NBT-272 and subjected to the same analysis. Results in the hit maps are expressed as the log2 ratio (where ratio is 2−ΔΔCt) obtained from NBT-272-treated or c-MYC siRNA-transfected cells normalized to the corresponding controls (i.e., mock-treated and control siRNA-transfected cells, respectively). Values in each analysis represent the average of 3 independent experiments.

Figure 4.

Indirect effect of NBT-272 on MYC. A, the effect of NBT-272 on either c-MYC or MYCN in different ET cell lines was analyzed at the protein (Western blotting) and at the mRNA level (qRT-PCR). B, cotreatment of D341 cells with MG132, inhibiting proteasome-mediated protein degradation, rescued c-MYC from the depleting effect of NBT-272. C, cell viability was evaluated in 3 NB-derived cell lines (SK-N-BE, SH-SY5Y, WAC-2) incubated for 72 hours with increasing doses of Mycro3 (34) or 10058-F4 (35). D, changes in the expression of apoptosis- (left) and cell cycle–related genes (right) were analyzed comparing the effect of NBT-272 (1) and of c-MYC downregulation by siRNA (2). For the silencing experiment, DAOY M2 cells were transfected transiently with c-MYC siRNA and gene expression was analyzed after 48 hours by using the RT2 Profiler PCR Array. The same cell line was treated for 24 hours with 20 nmol/L NBT-272 and subjected to the same analysis. Results in the hit maps are expressed as the log2 ratio (where ratio is 2−ΔΔCt) obtained from NBT-272-treated or c-MYC siRNA-transfected cells normalized to the corresponding controls (i.e., mock-treated and control siRNA-transfected cells, respectively). Values in each analysis represent the average of 3 independent experiments.

Close modal

The MYC family of transcription factors is known to have short half-lives (32). Therefore, we researched whether NBT-272 could have an effect on protein stability, thus helping to explain MYC depletion. In fact, by using the proteasome inhibitor MG132 (33) in combination with NBT-272, c-MYC was already almost completely rescued after 4 hours (Fig. 4B).

Additional lines of evidence supported an indirect effect of NBT-272 on MYC. Two small-molecule inhibitors specifically targeting the MYC/Max interaction (34, 35) did not show the same profile of activity as NBT-272 (Fig. 4C). In fact, significant cell viability could still be measured at high doses of both Mycro3 and 10058-F4. In particular, WAC-2 cells were almost unaffected by either of the compounds, whereas 103 times less concentrated NBT-272 completely arrested cellular growth.

Finally, only a limited number of gene expression changes induced by NBT-272 matched with the transcriptional effects of siRNA-mediated silencing of c-MYC in DAOY cells (Supplementary Fig. S6; Fig. 4D). In fact, only a restricted number of genes were up-/downregulated in both sets of experiments, that is, 11 apoptosis-related and 11 cell cycle–related transcripts (out of 84 in each analysis). Among them, even fewer genes were known direct targets of c-MYC (i.e., GADD45A, BIRC3, DFFA, CDK6, CDKN1A/p21, and PCNA). On the other hand, genes reported to be MYC targets exhibited a different expression profile, that is, they were mostly downregulated in c-MYC–depleted cells, but induced or left unchanged in cells treated with NBT-272 (e.g., the cyclins CCNB1, CCNB2, CCND2, CCNH, and the DNA replication regulators MCM4 and MCM5).

Altogether, these results led us to conclude that a direct inhibition of MYC is unlikely to explain the effects triggered by NBT-272 in ET cells, which are more likely to be the result of more complex mechanisms and of interference with upstream pathways.

NBT-272 targeted components of the AKT and ERK signaling pathways

To improve our understanding of the regulatory mechanisms leading to arrest of the cell cycle, apoptosis, and autophagy, we analyzed the activation status of 2 signaling pathways, that is, the AKT/mTOR and the MEK/ERK pathways, regulating these cellular responses (36). Furthermore, both pathways have been documented to be relevant also in the development of ET.

Figure 5A shows the effect of NBT-272 on the activation status of AKT varying in different cell lines, with phosphorylation at Ser473 significantly decreasing in WAC-2, SK-N-BE, and SH-SY5Y cell lines, but slightly enhanced in DAOY cells. On the other hand, the level of phosphorylated ERK 1/2 (p44/p42) diminished in all cell lines tested, although with different kinetics (Fig. 5A), suggesting a different sensitivity of the 2 pathways to NBT-272. To test whether NBT-272 had a direct effect on the enzymatic activity of either AKT or ERK and could thus be regarded as a kinase inhibitor, we quantified the in vitro kinase activity of 271 purified recombinant kinases (Supplementary Fig. S7A). However, none of them were significantly impaired by NBT-272 (20 nmol/L). Additionally, different subunits and isoforms of phosphoinositide-3 kinase (PI3K) were tested (Supplementary Fig. S7B), again showing no effect from NBT-272 (20 nmol/L).

Figure 5.

Inhibition of protein translation and of the AKT/mTOR- and ERK-dependent pathways. A, protein expression and activation status of components of the AKT/mTOR and ERK pathways were investigated by immunoblotting. B, treatment with NBT-272 triggered a depletion of active eIF4E and 4EBP-1 proteins in different cell lines as early as 16 hours.

Figure 5.

Inhibition of protein translation and of the AKT/mTOR- and ERK-dependent pathways. A, protein expression and activation status of components of the AKT/mTOR and ERK pathways were investigated by immunoblotting. B, treatment with NBT-272 triggered a depletion of active eIF4E and 4EBP-1 proteins in different cell lines as early as 16 hours.

Close modal

Although the exact mechanism of action of NBT-272 remained unsolved, the compound clearly affected AKT- and ERK-dependent cellular functions. The downstream target of ERK, MNK1 (37), was also deactivated on treatment with NBT-272, whereas the upstream kinase MEK1/2 was only slightly affected (Fig. 5A). MNK1 is known to phosphorylate both eIF4E and its repressor, 4EBP-1 (38). 4EBP-1 is also a direct target of mTOR, playing a key role as regulator of protein translation downstream of AKT (22). Interestingly, regardless of the AKT activation status, phosphorylation of both eIF4E and 4EBP-1 was impaired by NBT-272 in MB and NB cells (Fig. 5B), thus preventing the cap-dependent protein translation machinery from being completely functional. Additionally, the phosphorylation status of the ribosomal S6 protein, target of mTOR via S6 kinase (S6K; ref. 39), was proven to be unaffected, thus reinforcing the idea of a 4EBP-1–dependent block of protein synthesis.

Moreover, we asked whether the cellular sensitivity to NBT-272-mediated G1/S block of the cell cycle, apoptosis, or autophagy would vary if 1 of the pathways had been affected by using specific inhibitors. To test the relevance of the mTOR/AKT pathway, BEZ235 was used (40). A pretreatment with the PI3K/mTOR inhibitor increased both depletion of CCND1 and degradation of SQSTM1/p62 when compared with NBT-272 alone (Supplementary Fig. S8A), whereas no significant enhancement of the proapoptotic functions of NBT-272 was observed (Supplementary Fig. S8B). Although BEZ235 alone had apparently no effect, under these conditions, on the expression of the cell cycle and autophagy markers in DAOY cells, the inhibitor was functional, as shown by the strong deactivation of AKT (Supplementary Fig. S8A) and the induction of apoptosis (Supplementary Fig. S8B).

NBT-272 blocked tumor progression in a xenograft model of NB

NB cells bearing a transgene encoding luciferase (SH-SY5Y-LUC) were injected in the flank of 10 athymic nude mice and were allowed to develop for 3 weeks. The effect of the intraperitoneal administration of NBT-272 (10 μmol/L per week) and of the vehicle (0.003% EtOH/PBS) on the tumor growth was monitored by quantifying the luminescent signal from engrafted SH-SY5Y-LUC cells. After 6 weeks of treatment, the tumors almost stopped growing in the mice receiving NBT-272, whereas the human NB SH-SY5Y cells expanded into large tumors in the control animals (Fig. 6A and B; Supplementary Fig. S9). In fact, a 6.3-fold increase of tumor size could be detected in the control group compared with the NBT-272–treated group (P = 0.037 according to Student's t test). The result clearly indicated that NBT-272 has a significant antiproliferative effect on the tumor development in vivo. The hemotoxylin/eosin staining of some representative tumor samples showed a compact and rounded shape of the tumors, which were characterized by a very dense cell distribution with some necrotic areas (Fig. 6C, a–d). Tumors from NBT-272-treated mice showed a clear reduction of Ki67-positive cells (Fig. 6C, f and h), whereas a prominent cellular perinuclear staining in vehicle-treated mice was observed (Fig. 6C, e and g), thus confirming the significant antiproliferative effect of NBT-272 also in vivo. Activation of caspase-3 was also evaluated (Fig. 6C, i and j), which shows that the treatment with NBT-272 did not induce a significantly higher induction of apoptosis than with control tumors. This result confirmed our findings in vitro (Fig. 3A and B) by referring to the same cellular model, that is, SH-SY5Y cells, which were used to induce the tumors. Finally, NBT-272 treatment had an effect on MYC stability/expression in the tumors, in which ERK activation was also impaired (Fig. 6D).

Figure 6.

Reduction of tumor growth induced by NBT-272 in a xenograft model. Female athymic nude mice were injected in the right flank with SH-SY5Y-LUC cells. Three weeks after injection, the mice were divided into 2 homogeneous groups (based on the amount of engrafted human cells). The mice received either NBT-272 (10 μmol/L per week) or vehicle (0.003% EtOH/PBS), both administered 3 times a week. A, the tumor growth rate was monitored on a weekly basis by quantifying the luciferase activity of the recombinant tumor cells in vivo (see scale bars) and plotted starting from the first day of treatment. Blue, vehicle-treated animals. Red, NBT-272-treated animals. BLI data are available as a supplementary figure (see Supplementary Fig. 7S). B, quantification of the tumor growth rate. C, H&E and immunohistochemistry staining of tumor samples obtained from mice treated either with vehicle alone (left panels) or with NBT-272 (right panels) according to the described protocol. The panels c and d represent the enlarged view of the selected areas in panels a and b, respectively. In a and b, the tumors are shown as hyperdense-rounded structures with some necrosis portions (n). In panels e–h, the tumor sections were stained for Ki67 (proliferation), whereas in panels i and j the staining for cleaved caspase-3 (cl-CASP3, apoptosis) is shown. Images a and b, ×5 magnification; images c–j, ×40 magnification. D, Western blotting detection of c-MYC, p-ERK1/2, and p-AKT was done in total protein extracts from tumors developed by mice 4 (vehicle treated) and 9 (NBT-272 treated).

Figure 6.

Reduction of tumor growth induced by NBT-272 in a xenograft model. Female athymic nude mice were injected in the right flank with SH-SY5Y-LUC cells. Three weeks after injection, the mice were divided into 2 homogeneous groups (based on the amount of engrafted human cells). The mice received either NBT-272 (10 μmol/L per week) or vehicle (0.003% EtOH/PBS), both administered 3 times a week. A, the tumor growth rate was monitored on a weekly basis by quantifying the luciferase activity of the recombinant tumor cells in vivo (see scale bars) and plotted starting from the first day of treatment. Blue, vehicle-treated animals. Red, NBT-272-treated animals. BLI data are available as a supplementary figure (see Supplementary Fig. 7S). B, quantification of the tumor growth rate. C, H&E and immunohistochemistry staining of tumor samples obtained from mice treated either with vehicle alone (left panels) or with NBT-272 (right panels) according to the described protocol. The panels c and d represent the enlarged view of the selected areas in panels a and b, respectively. In a and b, the tumors are shown as hyperdense-rounded structures with some necrosis portions (n). In panels e–h, the tumor sections were stained for Ki67 (proliferation), whereas in panels i and j the staining for cleaved caspase-3 (cl-CASP3, apoptosis) is shown. Images a and b, ×5 magnification; images c–j, ×40 magnification. D, Western blotting detection of c-MYC, p-ERK1/2, and p-AKT was done in total protein extracts from tumors developed by mice 4 (vehicle treated) and 9 (NBT-272 treated).

Close modal

A fine regulation of protein turnover by a tunable modulation of protein stability requires specific post-translation modifications (such as phosphorylation) of key molecules. Many of these molecules are significant components of prosurvival pathways and often play a crucial role in tumor development and progression, in conditions of constitutive and growth factor–independent activation. In past decades, great efforts have been made to interfere with those signaling pathways by employing different strategies of targeted therapy, ranging from antibodies against receptor tyrosine kinases (RTK) to small-molecule inhibitors blocking downstream signaling effectors. Downstream of RTK activation, the induction of MYC transcription factors and of their pleiotropic effects on cell growth has been frequently associated with neoplastic transformation. Therefore, MYC itself clearly represents an attractive target for anticancer therapy (41), also due of its correlation with poor prognosis and high-grade malignancy in many types of tumors, including pediatric malignancies (10, 11, 42, 43).

In the context of MYC-overexpressing ET tumors, we wanted to investigate the biological responses of a group of ET-derived cell lines to NBT-272, a small molecule of about 550 Da, previously described as inhibitor of MYC (15, 16). Its toxicity and ability to prevent colony formation in vitro indicated a very strong effect of NBT-272 at low concentrations.

In the search for an association between toxicity of NBT-272 and MYC expression levels in different ET cell lines, no significant correlation could be found. This observation led us to question any direct effect of NBT-272 on the oncogene, which would more likely be the result of different mechanisms, such as interference with protein synthesis and stability, affecting also the MYC family members known to have short half-lives (32). Indeed, we could observe that inhibition of the proteasome-dependent protein degradation pathway restored the depleting effect of NBT-272 and almost completely stabilized c-MYC. Moreover, the effect induced by 2 specific MYC/Max inhibitors (34, 35) and by c-MYC silencing in ET cells did not match the cellular response to NBT-272, again detrimental to the hypothesis of a direct effect on MYC. This conclusion, however, certainly does not exclude that MYC inhibition contributed to the panel of cellular responses observed on NBT-272 treatment.

Even though the compound is not a direct kinase inhibitor, our study indicated the ability of NBT-272 to interfere with the activation status of different components of the AKT/mTOR and MEK/ERK signaling pathways. We therefore attributed the effect of NBT-272 on apoptosis, cell cycle, and autophagy to its ability to interfere with these pathways. Indeed, the basal level phosphorylation of ERK1/2, MNK1, and AKT in MB and NB cells was impaired by NBT-272, under the same conditions that induced efficient cell cycle arrest (G1/S) and autophagosome formation. It is also relevant to emphasize that the 2 pathways converge to regulate the activation status of eIF4E and 4EBP-1, both dephosphorylated (i.e., inactive) in the presence of NBT-272, thus leading us to speculate that targeting these pathways would prevent full activation of the cap-dependent protein translation machinery. Furthermore, MYC depletion could possibly be explained, not only as an effect of protein synthesis inhibition, but also as a consequence of decreased phosphorylation at its Ser62 residue, which has a stabilizing effect and is catalyzed by ERK (44).

In total, our observations strongly suggest that both the AKT and MEK/ERK signaling pathways participate in orchestrating the cellular responses to NBT-272, at least in vitro. Moreover, combination of NBT-272 with a specific inhibitor of PI3K/mTOR, BEZ235 (40), seemed to further increase the NBT-272–dependent effect on cell cycle and autophagy, but not apoptosis. Numerous components of the AKT/mTOR and MEK/ERK pathways have been found deregulated in cancer, including ET. High basal activation levels of AKT and of the S6 ribosomal protein were documented in MB (45, 46) and a relevant role in NB pathogenesis was attributed to PI3K, found to be overexpressed in NB tumors (47). Phosphorylation of AKT, S6, and ERK has also been reported as a frequent event in NB, in which AKT activation in particular was correlated with diverse indicators of aggressive disease and poor prognosis (48). These 2 signaling pathways were also negatively associated with malignancy in childhood rhabdomyosarcoma (49) and in Ewing's sarcoma (50), thus rendering it certainly worthwhile to promote the idea of developing common therapeutic strategies effective in different ET entities.

Finally, NBT-272 arrested tumor growth in a xenograft model of human NB, showing that the potent effect of this compound could also be reproduced in vivo, with concomitant reduction of MYC expression and of ERK activation in the treated tumors. These very encouraging results warrant the extension of investigations to more ET models.

In summary, NBT-272 induced a relatively complex pattern of cellular responses, including inhibition of crucial regulators of protein synthesis. All these events could be related functionally to interference with key cell survival pathways (AKT and MEK/ERK) playing a role in the pathogenesis of several ETs. Our findings increase the present level of understanding of the mechanisms of action of NBT-272, explaining its potent antitumor effect in in vitro and in vivo models of ET. This study certainly justifies further efforts to define more clearly the potential benefits of using NBT-272 in novel therapeutic strategies for pediatric tumors.

No potential conflicts of interest were disclosed.

We thank Dr. Brian Carter for his precious contribution in proofreading the manuscript and Dr. Beat Bornhauser and Prof. Shida Yousefi for helpful discussion. We are grateful to Lawrence Helson and James McChesney for providing NBT-272, developed by Tapestry Pharmaceuticals (Boulder, CO). We also thank Thererse Bruggmann and Gery Barmettler for their excellent technical support during the preparation and analysis of samples for electron microscopy.

Grant Support

Deborah Castelletti and Giulio Fiaschetti were supported by the European Community FP6, project STREP (EET-pipeline, number: 037260). Valeria Di Dato and Daniela De Martino were supported by FP6-EET pipeline LSH-CT-2006-037260 (M. Zollo), FP7-Tumic HEALTH-F2-2008-201662 (M. Zollo), Associazione italiana contro la lotta al Neuroblastoma “Progetto Pensiero” (M. Zollo), and AIRC Tumori Pediatrici 2007-2010 (M. Zollo).

1.
Scotting
PJ
,
Walker
DA
,
Perilongo
G
. 
Childhood solid tumours: a developmental disorder
.
Nat Rev Cancer
2005
;
5
:
481
8
.
2.
Grimmer
MR
,
Weiss
WA
. 
Childhood tumors of the nervous system as disorders of normal development
.
Curr Opin Pediatr
2006
;
18
:
634
8
.
3.
Guessous
F
,
Li
Y
,
Abounader
R
. 
Signaling pathways in medulloblastoma
.
J Cell Physiol
2008
;
217
:
577
83
.
4.
Packer
RJ
. 
Childhood brain tumors: accomplishments and ongoing challenges
.
J Child Neurol
2008
;
23
:
1122
7
.
5.
Schwab
M
,
Westermann
F
,
Hero
B
,
Berthold
F
. 
Neuroblastoma: biology and molecular and chromosomal pathology
.
Lancet Oncol
2003
;
4
:
472
80
.
6.
Maris
JM
. 
The biologic basis for neuroblastoma heterogeneity and risk stratification
.
Curr Opin Pediatr
2005
;
17
:
7
13
.
7.
Schwab
M
,
Ellison
J
,
Busch
M
,
Rosenau
W
,
Varmus
HE
,
Bishop
JM
. 
Enhanced expression of the human gene N-myc consequent to amplification of DNA may contribute to malignant progression of neuroblastoma
.
Proc Natl Acad Sci U S A
1984
;
81
:
4940
4
.
8.
Bordow
SB
,
Norris
MD
,
Haber
PS
,
Marshall
GM
,
Haber
M
. 
Prognostic significance of MYCN oncogene expression in childhood neuroblastoma
.
J Clin Oncol
1998
;
16
:
3286
94
.
9.
Grotzer
MA
,
Hogarty
MD
,
Janss
AJ
, et al
MYC messenger RNA expression predicts survival outcome in childhood primitive neuroectodermal tumor/medulloblastoma
.
Clin Cancer Res
2001
;
7
:
2425
33
.
10.
Pomeroy
SL
,
Tamayo
P
,
Gaasenbeck
M
, et al
Prediction of central nervous system embryonal tumour outcome based on gene expression
.
Nature
2002
;
415
:
436
42
.
11.
Stearns
D
,
Chaudhry
A
,
Abel
TW
,
Burger
PC
,
Dang
CV
,
Eberhart
CG
. 
c-Myc overexpression causes anaplasia in medulloblastoma
.
Cancer Res
2006
;
66
:
673
81
.
12.
Schmidt
EV
. 
The role of c-myc in cellular growth control
.
Oncogene
1999
;
18
:
2988
96
.
13.
Rosati
A
,
Quaranta
E
,
Ammirante
M
,
Turco
MC
,
Leone
A
,
De Feo
V
. 
Quassinoids can induce mitochondrial membrane depolarisation and caspase 3 activation in human cells
.
Cell Death Differ
2004
;
11
Suppl 2
:
S216
8
.
14.
Cuendet
M
,
Pezzuto
JM
. 
Antitumor activity of bruceantin: an old drug with new promise
.
J Nat Prod
2004
;
67
:
269
72
.
15.
Helson
L
,
McChesney
J
,
Bartyzel
P
. 
In vitro cytotoxic activity of NBT-272, a novel quassinoid analog
.
Geneva: Eur J Cancer
2004
;
2
Suppl; 2004
:
171
.
16.
von Bueren
AO
,
Shalaby
T
,
Rajtarova
J
, et al
Anti-proliferative activity of the quassinoid NBT-272 in childhood medulloblastoma cells
.
BMC Cancer
2007
;
7
:
19
.
17.
Fukamiya
N
,
Lee
KH
,
Muhammad
I
, et al
Structure-activity relationships of quassinoids for eukaryotic protein synthesis
.
Cancer Lett
2005
;
220
:
37
48
.
18.
Lachance
PE
,
Miron
M
,
Raught
B
,
Sonenberg
N
,
Lasko
P
. 
Phosphorylation of eukaryotic translation initiation factor 4E is critical for growth
.
Mol Cell Biol
2002
;
22
:
1656
63
.
19.
De Benedetti
A
,
Harris
AL
. 
eIF4E expression in tumors: its possible role in progression of malignancies
.
Int J Biochem Cell Biol
1999
;
31
:
59
72
.
20.
Bjornsti
MA
,
Houghton
PJ
. 
Lost in translation: dysregulation of cap-dependent translation and cancer
.
Cancer Cell
2004
;
5
:
519
23
.
21.
Pyronnet
S
. 
Phosphorylation of the cap-binding protein eIF4E by the MAPK-activated protein kinase MNK1
.
Biochem Pharmacol
2000
;
60
:
1237
43
.
22.
Sun
SY
,
Rosenberg
LM
,
Wang
X
, et al
Activation of AKT and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition
.
Cancer Res
2005
;
65
:
7052
8
.
23.
de Veas
R Garcia
,
Schweigerer
L
,
Medina
MA
. 
Modulation of the proteolytic balance plasminogen activator/plasminogen activator inhibitor by enhanced N-myc oncogene expression or application of genistein
.
Eur J Cancer
1998
;
34
:
1736
40
.
24.
Kumps
C, Van Roy N, Heyrman L, Goossens D, Del-Favero J, Noguera R
, et al
Multiplex Amplicon Quantification (MAQ), a fast and efficient method for the simultaneous detection of copy number alterations in neuroblastoma
.
BMC Genomics
2010
;
11
:
298
.
25.
Garzia
L
,
Andolfo
I
,
Cusanelli
E
, et al
MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in medulloblastoma
.
PLoS One
2009
;
4
:
e4998
.
26.
Van Den Heuvel
S
,
Harlow
E
. 
Distinct roles for cyclin-dependent kinases in cell cycle control
.
Science
1993
;
262
:
2050
4
.
27.
Harbour
JW
,
Luo
RX
,
Dei Santi
A
,
Postigo
AA
,
Dean
DC
. 
Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1
.
Cell
1999
;
98
:
859
69
.
28.
Tasdemir
E
,
Galluzzi
L
,
Maiuri
MC
, et al
Methods for assessing autophagy and autophagic cell death
.
Methods Mol Biol
2008
;
445
:
29
76
.
29.
Faivre
S
,
Kroemer
G
,
Raymond
E
. 
Current development of mTOR inhibitors as anticancer agents
.
Nat Rev Drug Discov
2006
;
5
:
671
88
.
30.
Ohsumi
Y
,
Mizushima
N
. 
Two ubiquitin-like conjugation systems essential for autophagy
.
Semin Cell Dev Biol
2004
;
15
:
231
6
.
31.
Pankiv
S
,
Clausen
TH
,
Lamark
T
, et al
p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy
.
J Biol Chem
2007
;
282
:
24131
45
.
32.
Sears
RC
. 
The life cycle of c-myc: from synthesis to degradation
.
Cell Cycle
2004
;
3
:
1133
7
.
33.
Kim
D
,
Kim
SH
,
Li
GC
. 
Proteasome inhibitors MG132 and lactacystin hyperphosphorylate HSF1 and induce hsp70 and hsp27 expression
.
Biochem Biophys Res Commun
1999
;
254
:
264
8
.
34.
Kiessling
A
,
Wiesinger
R
,
Sperl
B
,
Berg
T
. 
Selective inhibition of c-Myc/Max dimerization by a pyrazolo[1,5-a]pyrimidine
.
Chem Med Chem
2007
;
2
:
627
30
.
35.
Wang
H
,
Hammoudeh
DI
,
Follis
AV
, et al
Improved low molecular weight Myc-Max inhibitors
.
Mol Cancer Ther
2007
;
6
:
2399
408
.
36.
Song
G
,
Ouyang
G
,
Bao
S
. 
The activation of AKT/PKB signaling pathway and cell survival
.
J Cell Mol Med
2005
;
9
:
59
71
.
37.
O'Loghlen
A
,
Gonzalez
VM
,
Jurado
T
,
Salinas
M
,
Martin
ME
. 
Characterization of the activity of human MAP kinase-interacting kinase MNK1b
.
Biochim Biophys Acta
2007
;
1773
:
1416
27
.
38.
Richter
JD
,
Sonenberg
N
. 
Regulation of cap-dependent translation by eIF4E inhibitory proteins
.
Nature
2005
;
433
:
477
80
.
39.
Ali
SM
,
Sabatini
DM
. 
Structure of S6 kinase 1 determines whether raptor-mTOR or rictor-mTOR phosphorylates its hydrophobic motif site
.
J Biol Chem
2005
;
280
:
19445
8
.
40.
Maira
SM
,
Stauffer
F
,
Brueggen
J
, et al
Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity
.
Mol Cancer Ther
2008
;
7
:
1851
63
.
41.
Grotzer
MA
,
Castelletti
D
,
Fiaschetti
G
,
Shalaby
T
,
Arcaro
A
. 
Targeting Myc in pediatric malignancies of the central and peripheral nervous system
.
Curr Cancer Drug Targets
2009
;
9
:
176
88
.
42.
Rouah
E
,
Wilson
DR
,
Armstrong
DL
,
Darlington
GJ
. 
N-myc amplification and neuronal differentiation in human primitive neuroectodermal tumors of the central nervous system
.
Cancer Res
1989
;
49
:
1797
801
.
43.
Gilbertson
RJ
,
Wickramasinghe
C
,
Hernan
R
, et al
Clinical and molecular stratification of disease risk in medulloblastoma
.
Br J Cancer
2001
;
85
:
705
12
.
44.
Lutterbach
B
,
Hann
SR
. 
Hierarchical phosphorylation at N-terminal transformation-sensitive sites in c-Myc protein is regulated by mitogens and in mitosis
.
Mol Cell Biol
1994
;
14
:
5510
22
.
45.
Guerreiro
AS
,
Fattet
S
,
Fischer
B
, et al
Targeting the PI3K p110alpha isoform inhibits medulloblastoma proliferation, chemoresistance, and migration
.
Clin Cancer Res
2008
;
14
:
6761
9
.
46.
Hartmann
W
,
Digon-Sontgerath
B
,
Koch
A
, et al
Phosphatidylinositol 3′-kinase/AKT signaling is activated in medulloblastoma cell proliferation and is associated with reduced expression of PTEN
.
Clin Cancer Res
2006
;
12
:
3019
27
.
47.
Boller
D
,
Schramm
A
,
Doepfner
KT
, et al
Targeting the phosphoinositide 3-kinase isoform p110delta impairs growth and survival in neuroblastoma cells
.
Clin Cancer Res
2008
;
14
:
1172
81
.
48.
Opel
D
,
Poremba
C
,
Simon
T
,
Debatin
KM
,
Fulda
S
. 
Activation of AKT predicts poor outcome in neuroblastoma
.
Cancer Res
2007
;
67
:
735
45
.
49.
Petricoin
EF
 3rd
,
Espina
V
,
Araujo
RP
, et al
Phosphoprotein pathway mapping: AKT/mammalian target of rapamycin activation is negatively associated with childhood rhabdomyosarcoma survival
.
Cancer Res
2007
;
67
:
3431
40
.
50.
Benini
S
,
Manara
MC
,
Cerisano
V
, et al
Contribution of MEK/MAPK and PI3-K signaling pathway to the malignant behavior of Ewing's sarcoma cells: therapeutic prospects
.
Int J Cancer
2004
;
108
:
358
66
.

Supplementary data