Purpose: We aimed to identify novel prognostic biomarkers for Ewing's sarcoma by investigating the global protein expression profile of Ewing's sarcoma patients.

Experimental Design: We examined the proteomic profile of eight biopsy samples from Ewing's sarcoma patients using two-dimensional difference gel electrophoresis. Three patients were alive and continuously disease-free over 3 years after the initial diagnosis (good prognosis group) and five had died of the disease within 2 years of the initial diagnosis (poor prognosis group).

Results: The protein expression profiles produced using two-dimensional difference gel electrophoresis consisted of 2,364 protein spots, among which we identified 66 protein spots whose intensity showed >2-fold difference between the two patient groups. Mass spectrometric protein identification showed that the 66 spots corresponded to 53 distinct gene products. Pathway analysis revealed that 31 of 53 proteins, including nucleophosmin, were significantly related to bone tissue neoplasms (P < 0.000001). The prognostic performance of nucleophosmin was evaluated immunohistochemically on an additional 34 Ewing's sarcoma cases. Univariate and multivariate analyses revealed that nucleophosmin expression significantly correlated with overall survival (P < 0.01).

Conclusions: These results establish nucleophosmin as a candidate of independent prognostic marker for Ewing's sarcoma patients. Measuring nucleophosmin in biopsy samples before treatment may contribute to the effective management of Ewing's sarcoma.

Translational Relevance

In Ewing's sarcoma, a novel prognostic modality has long been desired to select the patients that would benefit from intensified treatment. We performed a proteomic study using incisionally biopsied samples before treatment. A comparative protein expression study in 8 patients identified nucleophosmin as a novel prognostic biomarker. A subsequent immunohistochemical study on a further 34 cases established the correlation between higher nucleophosmin expression and poor prognosis. In our study, nucleophosmin was identified as a novel candidate prognostic biomarker through the use of modern global protein expression modalities. Our study suggests the possible use of nucleophosmin expression for personalized medicine for Ewing's sarcoma patients.

Ewing's sarcoma is the second most common primary malignant bone tumor in children and adolescents. Despite significant progress regarding intensive chemotherapy protocols and local control measures, 30% to 40% of patients with localized Ewing's sarcoma and 80% of patients with metastatic Ewing's sarcoma at diagnosis die due to disease progression within 5 years (1). More intensified first-line chemotherapy regimens or combinations of chemotherapeutic agents improve clinical outcome compared with conventional chemotherapy (2, 3). However, such therapies may result in serious toxicity, including fatal gastrointestinal toxicity, grade 3 or 4 infections, and severe myelosuppression (46). The patients could thus benefit from less aggressive regimens by avoiding the higher risk of toxicity associated with overtreatment. Indeed, approximately two-thirds of patients with localized Ewing's sarcoma are cured with conventional therapy alone (7, 8). Therefore, the identification of prognostic factors may lead to the development of risk-adapted treatment strategies for Ewing's sarcoma.

Clinical factors currently evaluated have limited prognostic value; the presence of metastases at diagnosis, which is the most unfavorable prognostic factor for Ewing's sarcoma, concerns only ∼25% of Ewing's sarcoma patients (9). The prognostic value of other clinical and pathologic features that correlated with prognosis of Ewing's sarcoma, including the site and size of the lesion and the age of the patient (1, 10), has decreased following recent advances in treatment (11). For instance, in earlier studies, tumors >8 cm were associated with a worse prognosis (12), whereas tumor size is not assumed as definitive prognostic factor in studies using the more intensive EW92 protocol (13).

In recent years, high-throughput screening technologies such as array-based comparative genomic hybridization analysis and cDNA microarray technology have been used to identify up-regulated or down-regulated genes with prognostic value for Ewing's sarcoma (1419). These comprehensive studies suggested the presence of a poor prognosis signature at diagnosis and identified several genes that may be involved in the process of invasion and metastasis in Ewing's sarcoma.

Emerging technologies that examine the overall features of the expressed proteins, that is, proteomics, have identified many candidate proteins associated with early diagnosis (20), differential diagnosis (21), prognosis (22, 23), and response to chemotherapy (24) in various diseases but have not been vigorously employed in the study of Ewing's sarcoma.

In this study, we performed a proteomic study using biopsy samples from Ewing's sarcoma patients. We found that nucleophosmin expression significantly correlated with progression of Ewing's sarcoma. Although aberrant expression of nucleophosmin has been implicated in various other malignancies (2529), this proteomic study shows its aberrant expression and prognostic utility in Ewing's sarcoma.

Patients and clinical information. This study included a total of eight frozen incisional biopsy samples taken before treatment at the time of diagnosis from 8 Ewing's sarcoma patients treated between June 1996 and December 2006. These samples were snap-frozen in liquid nitrogen and stored at −80°C until use. The clinical information of the patients is summarized in Table 1. This project was approved by the ethical review board of the National Cancer Center after signed informed consent was obtained from all patients. All cases were reviewed and histopathologically diagnosed by a certified pathologist (N.T. and T.S.). Clinical staging was determined based on diagnostic imaging criteria according to the Musculoskeletal Tumor Society Surgical staging system (30). Primary tumor size was measured at the greatest tumor dimension on radiographic images, including computed tomography scans and magnetic resonance imaging.

Table 1.

Clinicopathologic features of the cases, the frozen samples of which were examined by proteomics

Case no.Age/sex(y)Primary siteSize (cm)Sample sourceStage*Metastatic site (first development)Metastasis time after diagnosis (mo)Follow-up period after diagnosis (mo)Follow-up status
M/19 Thigh Biopsy None None 88 CDF 
F/18 Chest wall Biopsy None None 70 CDF 
F/9 Parietal Bone Biopsy None None 46 CDF 
M/62 Thigh Biopsy Lymph node, Brain At diagnosis 12 DOD 
F/28 Humerus Biopsy Lung DOD 
M/32 Thigh 15 Biopsy Lung At diagnosis 10 DOD 
M/12 Ilium 11 Biopsy Bone DOD 
Case no.Age/sex(y)Primary siteSize (cm)Sample sourceStage*Metastatic site (first development)Metastasis time after diagnosis (mo)Follow-up period after diagnosis (mo)Follow-up status
M/19 Thigh Biopsy None None 88 CDF 
F/18 Chest wall Biopsy None None 70 CDF 
F/9 Parietal Bone Biopsy None None 46 CDF 
M/62 Thigh Biopsy Lymph node, Brain At diagnosis 12 DOD 
F/28 Humerus Biopsy Lung DOD 
M/32 Thigh 15 Biopsy Lung At diagnosis 10 DOD 
M/12 Ilium 11 Biopsy Bone DOD 
*

Stages I and II defined as localized disease (L) and stage III defined as metastatic disease (M).

From the 8 cases included in the study, 3 patients were alive and continuously disease-free (CDF) in the follow-up period of at least 3 years from diagnosis and 5 patients were dead of disease (DOD) within 2 years from initial diagnosis. All 5 patients in the latter group developed distant metastases within 7 months from initial diagnosis.

A previous report indicated that Ewing's sarcoma patients with early relapse, defined as relapse within 2 years after initial diagnosis, had shorter survival (31). We grouped the Ewing's sarcoma samples into two groups: the samples from patients that were alive and CDF over 3 years post-diagnosis were defined as the good prognosis group (Table 1, samples 1-3). The samples from patients that were DOD within 2 years were defined as the poor prognosis group (Table 1, samples 4-8).

For the nucleophosmin immunohistochemical expression study, we examined 34 tissues paraffin-embedded before treatment from 34 independent cases (Table 2, samples 9-42). These patients were treated between June 1981 and December 2005 at the National Cancer Center and Keio University Hospital. This project was approved by the ethical review boards of the National Cancer Center and Keio University Hospital after signed informed consent was obtained from all patients in this study. The clinical information concerning the cases used in the immunohistochemical study is summarized in Table 2.

Table 2.

Clinicopathologic features of the 34 Ewing's sarcoma cases examined immunohistochemically

Case no.Sex/age (y)Primary siteSize (cm)Sample sourceStage*Metastatic site (first development)Metastasis time after diagnosis (mo)Follow-up period after diagnosis (mo)Follow-up statusNucleophosmin positivityFusion geneChemotherapyChemotherapy agentsOperationRadiation
M/9 Fibula Biopsy None None 93 CDF − NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc Amputation − 
10 M/14 Clavicle 15 Biopsy None None 177 CDF − NT VAIA VCR, ACT, IFO, DOX Wide resection 
11 M/49 Femur 11 Biopsy None None 141 CDF − NT VAIA VCR, ACT, IFO, DOX Wide resection − 
12 M/16 Arm Biopsy None None 93 CDF − NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc Wide resection − 
13 M/25 Rib 5.5 Biopsy None None 75 CDF − NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc Wide resection − 
14 M/9 Talus Biopsy None None 70 CDF NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc − 
15 M/1 Tibia  Biopsy None None 179 CDF NT VAC VCR, ACT, CYC Amputation 
16 F/36 Femur  Biopsy None None 174 CDF − NT VACA VCR, ACT, CYC, DOX Amputation 
17 M/13 Tibia  Biopsy None None 166 CDF − EWS/ERG CYVADIC CYC, VCR, DOX, DTIC Wide resection 
18 F/22 Fibula Biopsy None None 108 CDF − EWS/FLI1 type 2 KS-1 ETO, CDDP, THP, IFO Wide resection − 
19 M/18 Rib 6.5 Biopsy None None 105 CDF − EWS/FLI1 EWSex10/FLI-1ex6 KS-1 ETO, CDDP, THP, IFO Wide resection 
20 M/35 Thigh Biopsy None None 126 CDF NT KS-1 ETO, CDDP, THP, IFO Wide resection − 
21 F/36 Arm Biopsy None None 101 CDF Not detected KS-1 ETO, CDDP, THP, IFO Wide resection − 
22 M/15 Thigh 16 Biopsy Lung 11 DOD NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO Wide resection 
23 M/17 Rib 10 Biopsy Multiple bone DOD EWS R1 rearrangement ACT + CDDP ACT, CDDP − 
24 M/18 Back 25 Biopsy Lung DOD NT VACA VCR, ACT, CYC, DOX Intralesional resection 
25 M/22 Femur 10 Biopsy Bone 14 DOD NT VAIA VCR, ACT, IFO, DOX − − 
26 M/37 Ilium 25 Biopsy Lung At diagnosis 22 DOD − NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO − 
27 F/24 Sacrum 10 Biopsy Bone 22 DOD NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO − 
28 M/11 Femur 10 Biopsy Bone, brain, lung 32 DOD NT VAC VCR, ACT, CYC Amputation 
29 F/22 Humerus 18 Biopsy Bone 19 32 DOD NT VAIA VCR, ACT, IFO, DOX Wide resection 
30 F/20 Femur Biopsy Lung 41 75 DOD NT T11 CYC, DOX,MTX, VCR + BLM, CYC, ACT + CYC, DOX, MTX Wide resection − 
31 M/21 Parietal bone Biopsy Lung 69 94 DOD NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO − 
32 F/19 Humerus Biopsy Lung, bone 43 121 DOD NT T11 CYC, DOX,MTX, VCR + BLM, CYC, ACT + CYC, DOX, MTX − 
33 M/17 Vertebra 10 Biopsy Lung 12 45 DOD NT T11 CYC, DOX,MTX, VCR + BLM, CYC, ACT + CYC, DOX, MTX Marginal resection 
34 F/16 Tibia 20 Biopsy Lung At diagnosis 16 DOD EWS/FLI1 type 2 KS-1 ETO, CDDP, THP, IFO Wide resection − 
35 M/18 Fibula Biopsy Bone, lung 21 DOD EWS/FLI1 type 1 KS-1 ETO, CDDP, THP, IFO Wide resection 
36 M/23 Pelvis 13 Biopsy Lung At diagnosis 17 DOD NT KS-1 ETO, CDDP, THP, IFO − 
37 M/29 Thigh 16 Biopsy Chest At diagnosis 12 DOD EWS/FLI1 type 2 KS-1 ETO, CDDP, THP, IFO Intralesional resection 
38 F/63 Paravertebra 17 Biopsy None None 71 DOD NT KS-1 ETO, CDDP, THP, IFO Intralesional resection 
39 F/20 Lower leg 10 Biopsy Lung, lymph node 14 DOD − EWS/FLI1 type 1 KS-1 ETO, CDDP, THP, IFO Marginal resection 
40 F/56 Forearm Biopsy Lung At diagnosis 11 DOD NT KS-1 ETO, CDDP, THP, IFO Wide resection − 
41 M/7 Paravertebra Biopsy Brain 15 16 DOD Not detected KS-1 ETO, CDDP, THP, IFO Marginal resection − 
42 F/11 Paravertebra Biopsy Lung 17 22 DOD EWS/FLI1 type 1 KS-1 ETO, CDDP, THP, IFO Intralesional resection 
Case no.Sex/age (y)Primary siteSize (cm)Sample sourceStage*Metastatic site (first development)Metastasis time after diagnosis (mo)Follow-up period after diagnosis (mo)Follow-up statusNucleophosmin positivityFusion geneChemotherapyChemotherapy agentsOperationRadiation
M/9 Fibula Biopsy None None 93 CDF − NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc Amputation − 
10 M/14 Clavicle 15 Biopsy None None 177 CDF − NT VAIA VCR, ACT, IFO, DOX Wide resection 
11 M/49 Femur 11 Biopsy None None 141 CDF − NT VAIA VCR, ACT, IFO, DOX Wide resection − 
12 M/16 Arm Biopsy None None 93 CDF − NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc Wide resection − 
13 M/25 Rib 5.5 Biopsy None None 75 CDF − NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc Wide resection − 
14 M/9 Talus Biopsy None None 70 CDF NT VCD + I-based regimens VCR, CYC, DOX, IFO, etc − 
15 M/1 Tibia  Biopsy None None 179 CDF NT VAC VCR, ACT, CYC Amputation 
16 F/36 Femur  Biopsy None None 174 CDF − NT VACA VCR, ACT, CYC, DOX Amputation 
17 M/13 Tibia  Biopsy None None 166 CDF − EWS/ERG CYVADIC CYC, VCR, DOX, DTIC Wide resection 
18 F/22 Fibula Biopsy None None 108 CDF − EWS/FLI1 type 2 KS-1 ETO, CDDP, THP, IFO Wide resection − 
19 M/18 Rib 6.5 Biopsy None None 105 CDF − EWS/FLI1 EWSex10/FLI-1ex6 KS-1 ETO, CDDP, THP, IFO Wide resection 
20 M/35 Thigh Biopsy None None 126 CDF NT KS-1 ETO, CDDP, THP, IFO Wide resection − 
21 F/36 Arm Biopsy None None 101 CDF Not detected KS-1 ETO, CDDP, THP, IFO Wide resection − 
22 M/15 Thigh 16 Biopsy Lung 11 DOD NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO Wide resection 
23 M/17 Rib 10 Biopsy Multiple bone DOD EWS R1 rearrangement ACT + CDDP ACT, CDDP − 
24 M/18 Back 25 Biopsy Lung DOD NT VACA VCR, ACT, CYC, DOX Intralesional resection 
25 M/22 Femur 10 Biopsy Bone 14 DOD NT VAIA VCR, ACT, IFO, DOX − − 
26 M/37 Ilium 25 Biopsy Lung At diagnosis 22 DOD − NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO − 
27 F/24 Sacrum 10 Biopsy Bone 22 DOD NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO − 
28 M/11 Femur 10 Biopsy Bone, brain, lung 32 DOD NT VAC VCR, ACT, CYC Amputation 
29 F/22 Humerus 18 Biopsy Bone 19 32 DOD NT VAIA VCR, ACT, IFO, DOX Wide resection 
30 F/20 Femur Biopsy Lung 41 75 DOD NT T11 CYC, DOX,MTX, VCR + BLM, CYC, ACT + CYC, DOX, MTX Wide resection − 
31 M/21 Parietal bone Biopsy Lung 69 94 DOD NT National Cancer Institute protocol (VAC + IE) VCR, DOX,CYC + IFO, ETO − 
32 F/19 Humerus Biopsy Lung, bone 43 121 DOD NT T11 CYC, DOX,MTX, VCR + BLM, CYC, ACT + CYC, DOX, MTX − 
33 M/17 Vertebra 10 Biopsy Lung 12 45 DOD NT T11 CYC, DOX,MTX, VCR + BLM, CYC, ACT + CYC, DOX, MTX Marginal resection 
34 F/16 Tibia 20 Biopsy Lung At diagnosis 16 DOD EWS/FLI1 type 2 KS-1 ETO, CDDP, THP, IFO Wide resection − 
35 M/18 Fibula Biopsy Bone, lung 21 DOD EWS/FLI1 type 1 KS-1 ETO, CDDP, THP, IFO Wide resection 
36 M/23 Pelvis 13 Biopsy Lung At diagnosis 17 DOD NT KS-1 ETO, CDDP, THP, IFO − 
37 M/29 Thigh 16 Biopsy Chest At diagnosis 12 DOD EWS/FLI1 type 2 KS-1 ETO, CDDP, THP, IFO Intralesional resection 
38 F/63 Paravertebra 17 Biopsy None None 71 DOD NT KS-1 ETO, CDDP, THP, IFO Intralesional resection 
39 F/20 Lower leg 10 Biopsy Lung, lymph node 14 DOD − EWS/FLI1 type 1 KS-1 ETO, CDDP, THP, IFO Marginal resection 
40 F/56 Forearm Biopsy Lung At diagnosis 11 DOD NT KS-1 ETO, CDDP, THP, IFO Wide resection − 
41 M/7 Paravertebra Biopsy Brain 15 16 DOD Not detected KS-1 ETO, CDDP, THP, IFO Marginal resection − 
42 F/11 Paravertebra Biopsy Lung 17 22 DOD EWS/FLI1 type 1 KS-1 ETO, CDDP, THP, IFO Intralesional resection 

NOTE: Chemotherapy agents: VACD, vincristine (VCR), actinomycin D (ACT), cyclophosphamide (CYC), and doxorubicin (DOX); IE, ifosfamide (IFO) and etoposide (ETO); THP, therarubicin; CDDP, cisplatin; BLM, bleomycin; MTX, methotrexate; DTIC, dacarbazine. NT, not tested.

*

Stages I and II defined as localized disease (L) and stage III defined as metastatic disease (M).

Tumor size cannot be evaluated.

Rearrangement analysis. Total RNA from tumors was extracted by the guanidinium thiocyanate method (ISOGEN, Nippon Gene). Samples were ground in a microcentrifuge tube. cDNA was generated using a first-strand cDNA synthesis kit (Pharmacia Biotech). Total RNA (1-5 μg) was transcribed. PCR was carried out in a 100 mL reaction mixture containing 1 to 7 μL cDNA template, 200 mmol/L deoxynucleotide triphosphates, 0.5 mmol/L of each oligonucleotide primer, and 2.5 units Taq polymerase in a 10 mmol/L Tris-HCl (pH 8.8) containing 50 mmol/L KCl and 1.5 mmol/L MgCl2. The oligonucleotide primers used for the PCR were ESBP-1 (EWS specific), ESBP-2 (FLI-1 specific), and primers specific for ERG, E1AF, and ETV1 (32). PCR was done in 35 cycles under the following protocol: denaturation at 94°C for 1 min, annealing at 65°C for 1 min, and elongation at 72°C for 1 min. The amplified products were visualized on 1% agarose gels.

Protein expression profiling. Frozen samples were crushed to powder with a Multi-beads shocker (Yasui Kikai) with liquid nitrogen. The frozen powder was then treated with urea lysis buffer (6 mol/L urea, 2 mol/L thiourea, 3% CHAPS, 1% Triton X-100). After centrifugation at 15,000 rpm for 30 min, the supernatant was recovered and used in the subsequent protein expression studies.

Two-dimensional difference gel electrophoresis was done as described previously (33). In brief, the internal control sample was prepared by mixing a portion of all individual samples. Five micrograms of the internal control sample and of each individual sample were labeled with Cy3 and Cy5, respectively (CyDye DIGE Fluor saturation dye; GE Healthcare Biosciences) according to the manufacturer's instructions. The differently labeled protein samples were mixed and separated by two-dimensional difference gel electrophoresis. The first dimension separation was achieved using IPG DryStrip gels (24 cm length, pI range between 4 and 7; GE Healthcare Biosciences). The second dimension separation was achieved by SDS-PAGE on large-format gels (38 cm length, Bio-craft, Itabashi; ref. 33). The gels were scanned using laser scanners (Typhoon Trio; GE Healthcare Biosciences) at appropriate wavelengths (Fig. 1A). For all spots, the intensity of the Cy5 image was normalized by that of the Cy3 image in the identical gel so that gel-to-gel differences were compensated using the Progenesis PG240 software (Nonlinear Dynamics). System reproducibility was verified by comparing the protein profiles obtained from three independent separations of the same sample (Table 1, case 1). Scatter plot analysis revealed that the standardized intensity of >96.6% of the spots ranged within a 2-fold difference (R = 0.9103; Fig. 1B).

Fig. 1.

Identification of proteins differentially expressed in Ewing's sarcoma. A, schematic workflow of sample preparation for quantitative analysis. Protein lysates are labeled with fluorescent dyes of different wavelengths of excitation and emission. Cy3-labeled samples are simultaneously mixed and divided into Cy5-labeled samples. Then, mixture of Cy3- and Cy5-labeled lysates are coseparated by two-dimensional difference gel electrophoresis. The gel is scanned with two wavelengths, each specific either for Cy3 or Cy5 dye. B, scattergram of expression profile of Ewing's sarcoma tissues. Comparison of data from three independent experiments revealed the high reproducibility of protein expression profiling. C, representative two-dimensional image of proteins detected in Ewing's sarcoma tissues. The 66 spots identified in this study are circled and numbered. The spot numbers correspond to those in Fig. 2 and Supplementary Table S1.

Fig. 1.

Identification of proteins differentially expressed in Ewing's sarcoma. A, schematic workflow of sample preparation for quantitative analysis. Protein lysates are labeled with fluorescent dyes of different wavelengths of excitation and emission. Cy3-labeled samples are simultaneously mixed and divided into Cy5-labeled samples. Then, mixture of Cy3- and Cy5-labeled lysates are coseparated by two-dimensional difference gel electrophoresis. The gel is scanned with two wavelengths, each specific either for Cy3 or Cy5 dye. B, scattergram of expression profile of Ewing's sarcoma tissues. Comparison of data from three independent experiments revealed the high reproducibility of protein expression profiling. C, representative two-dimensional image of proteins detected in Ewing's sarcoma tissues. The 66 spots identified in this study are circled and numbered. The spot numbers correspond to those in Fig. 2 and Supplementary Table S1.

Close modal

Protein identification by mass spectrometry. The proteins corresponding to the spots detected were identified using mass spectrometry according to our previous report (33). In brief, 100 μg Cy5- or Cy3-labeled proteins were separated by two-dimensional PAGE, recovered as gel plugs, and digested with modified trypsin (Promega). The trypsin digests were subjected to liquid chromatography (Paradigm MS4 dual solvent delivery system; Michrom BioResources) and mass spectrometry using a Finnigan LTQ linear ion trap mass spectrometer (Thermo Electron) equipped with a nano-electrospray ion source (AMR, Megro). The Mascot software (version 2.1; Matrix Science) was used to search for the mass of the peptide ion peaks against the SWISS-PROT database (Homo sapiens, 12867 sequence in Sprot_47.8 fasta file).

Functional classification of the identified proteins. Functional classification of the identified proteins was carried out according to their classification in Gene Ontology.6

Pathway analysis. Pathway analysis of the identified proteins was done using the MetaCore software analysis tool (GeneGo). MetaCore identifies networks based on a manually curated database containing known molecular interactions, functions, and disease interrelationships using proteome data. The pathways were identified by the probability that a random set of proteins with the same size as the input list would give rise to a particular mapping by chance. The identified networks were traced using the Metacore pre-filter tool. The Disease tab tool was used to automatically trace key proteins associated with disease networks stored in Metacore and to list the P value for each disease listed.

Immunohistochemical study. Nucleophosmin expression was examined immunohistochemically on paraffin-embedded tissues. In brief, 4-μm-thick tissue sections were autoclaved in 10 mmol/L citrate buffer (pH 6.0) at 121°C for 30 min and incubated with an antibody against nucleophosmin (sc-53175; Santa Cruz Biotechnology; 1:500 dilution) at room temperature. Immunostaining was done using the Envision Plus detection system (DAKO). Two observers (N.T. and K.K.) evaluated the staining in a blinded fashion for clinical data.

Statistical analysis. Hierarchical clustering was done using the Expressionist software (Genedata).

Statistical computations were done using the StatView version 5.0 statistical package (SAS Institute). Survival time was defined as the period from diagnosis to last follow-up (or death). Survival rate was estimated by the Kaplan-Meier method (34). The relationship between survival and other variables was investigated using the log-rank test for categorical variables and a score test based on the Cox proportional hazards model for continuous variables. A multivariate model was fitted using Cox regression with significant variables at the univariate level (P < 0.01; ref. 35). Following a large-scale cooperative study by the Japanese Musculoskeletal Oncology Group (36), in which age <16 years and tumor size <10 cm were shown to have a significantly worse clinical outcome by univariate survival analysis, we selected these cutoff values for this analysis.

We generated and compared the protein expression profiles between three good prognosis and five poor prognosis Ewing's sarcoma cases using two-dimensional difference gel electrophoresis. We detected 2,364 protein spots that appeared in all the images of the Cy3-labeled internal control sample. Among these 2,364 spots, 66 showed significantly (>2-fold ratio of means) different intensity between the two groups. The localization of the 66 spots on the two-dimensional image is shown in Fig. 1C. Using hierarchical clustering, the 66 spots were classified into two major groups, cluster A (7 spots) and cluster B (59 spots; Fig. 2). The intensity of the 7 spots belonging to cluster A was decreased, whereas that of the 59 spots of cluster B was increased in the poor prognosis group.

Fig. 2.

Hierarchical clustering of the 8 Ewing's sarcoma cases based on the intensity of the 66 protein spots detected. The cases are color-coded as yellow (good prognosis group) or light blue (poor prognosis group). The spot numbers, protein names, Gene Ontology (functional classification), and disease (bone tissue neoplasms; color-coded as red) related proteins are shown (right).

Fig. 2.

Hierarchical clustering of the 8 Ewing's sarcoma cases based on the intensity of the 66 protein spots detected. The cases are color-coded as yellow (good prognosis group) or light blue (poor prognosis group). The spot numbers, protein names, Gene Ontology (functional classification), and disease (bone tissue neoplasms; color-coded as red) related proteins are shown (right).

Close modal

Mass spectrometric analysis resulted in the identification of 53 distinct gene products (6 proteins in cluster A and 47 proteins in cluster B) corresponding to the 66 protein spots (Fig. 2; Supplementary Table S1).

Although all six proteins in cluster A belonged to different functional categories as classified in Gene Ontology, most of the proteins in cluster B were divided into eight main categories: cytoskeletal/structural protein, transcription/translation, signal transduction, transport, antiapoptosis, response oxidative stress, acute-phase response, and cell proliferation (Fig. 2).

We further explored the biological significance of the altered protein expression pattern in cluster B based on a manually curated database containing known molecular interactions, functions, and disease interrelationships using MetaCore. We found that 31 (65.9%) of 47 proteins were functionally linked with each other and that the identified network of the 31 proteins was significantly related to bone tissue neoplasms (P < 0.000001; Fig. 2).

In the 31 proteins of cluster B, nucleophosmin was included. Although aberrant expression of nucleophosmin has been implicated in various other malignancies (2529), its association with Ewing's sarcoma has not been reported previously. Therefore, we validated the correlation of nucleophosmin with prognosis using immunohistochemistry in an additional 34 Ewing's sarcoma cases. Two patterns of nucleophosmin-positive staining were observed, both nuclear: a dot-like pattern and a diffuse-like pattern (Fig. 3). Similar to previous reports (25, 37), cases with nuclear staining for nucleophosmin were considered as nucleophosmin positive (23 of 34 cases; Table 2), whereas cases without staining for nucleophosmin were considered as nucleophosmin negative.

Fig. 3.

Nucleophosmin expression was observed in Ewing's sarcoma by immunohistochemistry. A to C, H&E staining of Ewing's sarcoma cases. Strongly stained nucleophosmin nuclear expression was observed in tumor cells. D, dot-like pattern staining. E, diffuse-like pattern staining. F, not observed stained nucleophosmin nuclear expression. Case numbers correspond to those in Table 2. Magnification, ×400.

Fig. 3.

Nucleophosmin expression was observed in Ewing's sarcoma by immunohistochemistry. A to C, H&E staining of Ewing's sarcoma cases. Strongly stained nucleophosmin nuclear expression was observed in tumor cells. D, dot-like pattern staining. E, diffuse-like pattern staining. F, not observed stained nucleophosmin nuclear expression. Case numbers correspond to those in Table 2. Magnification, ×400.

Close modal

In the follow-up period (median, 57.5 months; range, 8-179 months), 13 of 34 patients were alive and CDF and 21 patients were DOD (Fig. 4A).

Fig. 4.

Kaplan-Meier estimated overall survival curves are illustrated for all patients (n = 34; A), for all patients based on nucleophosmin positivity (B), and for patients with localized disease (n = 29) by nucleophosmin positivity (C). Statistically significant differences in overall survival periods were observed between the nucleophosmin-positive and nucleophosmin-negative cases both for all cases (P < 0.01, log-rank test) and the cases with localized disease (P < 0.01, log-rank test).

Fig. 4.

Kaplan-Meier estimated overall survival curves are illustrated for all patients (n = 34; A), for all patients based on nucleophosmin positivity (B), and for patients with localized disease (n = 29) by nucleophosmin positivity (C). Statistically significant differences in overall survival periods were observed between the nucleophosmin-positive and nucleophosmin-negative cases both for all cases (P < 0.01, log-rank test) and the cases with localized disease (P < 0.01, log-rank test).

Close modal

The group of nucleophosmin-positive cases included a significantly higher number of DOD patients compared with the nucleophosmin-negative group (P < 0.01, log-rank test; Fig. 4B), showing that nucleophosmin expression correlates with prognosis.

Following univariate analysis, nucleophosmin positivity (P < 0.01) and clinical stage (presence of metastatic disease at diagnosis; P < 0.01) significantly correlated with shorter overall survival. No other factors examined, including the tumor size, age at diagnosis, sex, chemotherapy regimens, tumor resectibility, and primary site, were associated with overall survival (Table 3).

Table 3.

Univariate and multivariate analyses of prognostic factors

VariableUnivariate survival analysis
Multivariate survival analysis
No. casesNo. aliveUnivariate PRisk ratio (95% confidence interval)PRelative risk (95% confidence interval)
Age at diagnosis (y)       
    <16 0.3209   
    ≥16 25  1.738 (0.583-5.180)   
Sex       
    M 22 10 0.5325   
    F 12  0.517 (0.207-1.288)   
Primary site       
    Extremity 21 10 0.212   
    Axial 13  1.736 (0.730-4.126)   
Tumor size (cm)*       
    <10 16 0.0182   
    ≥10 15  2.952 (1.202-7.251)   
Clinical stage       
    Localized 29 13 <0.01 0.0039 
    Metastatic  5.238 (1.697-16.164)  5.964 (1.773-20.060) 
Chemotherapy regimens       
    Including IE 17 0.0629   
    Not including IE 17  0.425 (0.955-5.791)   
Tumor resectibility       
    Resectable 26 12 0.1219   
    Nonresectable  2.004 (0.200-1.244)   
Nucleophosmin immunohistochemistry       
    Negative 11 <0.01 0.0063 
    Positive 23  7.425 (1.715-32.147)  7.768 (1.783-33.841) 
VariableUnivariate survival analysis
Multivariate survival analysis
No. casesNo. aliveUnivariate PRisk ratio (95% confidence interval)PRelative risk (95% confidence interval)
Age at diagnosis (y)       
    <16 0.3209   
    ≥16 25  1.738 (0.583-5.180)   
Sex       
    M 22 10 0.5325   
    F 12  0.517 (0.207-1.288)   
Primary site       
    Extremity 21 10 0.212   
    Axial 13  1.736 (0.730-4.126)   
Tumor size (cm)*       
    <10 16 0.0182   
    ≥10 15  2.952 (1.202-7.251)   
Clinical stage       
    Localized 29 13 <0.01 0.0039 
    Metastatic  5.238 (1.697-16.164)  5.964 (1.773-20.060) 
Chemotherapy regimens       
    Including IE 17 0.0629   
    Not including IE 17  0.425 (0.955-5.791)   
Tumor resectibility       
    Resectable 26 12 0.1219   
    Nonresectable  2.004 (0.200-1.244)   
Nucleophosmin immunohistochemistry       
    Negative 11 <0.01 0.0063 
    Positive 23  7.425 (1.715-32.147)  7.768 (1.783-33.841) 
*

Tumor size could not be evaluated in 3 Ewing's sarcoma cases.

We investigated whether nucleophosmin expression significantly correlated with the original tumor site and the tumor respectability status. Of the 34 cases in this study, 21 originated at an extra-axial and 13 at an axial site. Nucleophosmin expression did not correlate with the original tumor site (P = 0.87, Fisher's test). Of the 21 extra-axial tumors, 14 were nucleophosmin positive, and nucleophosmin expression correlated with poor prognosis (P < 0.05, log-rank test). Of the 13 axial tumors, 9 were nucleophosmin negative; again, nucleophosmin expression significantly correlated with poor prognosis (P < 0.01, log-rank test). Therefore, nucleophosmin expression correlated with poor prognosis independent of the original tumor site.

There was no significant difference regarding the prognosis between the resectable (26 of 34) and nonresectable cases (P = 0.1219, log-rank test). Among the resectable tumors, 16 were nucleophosmin positive and had worse prognosis than the remaining 10 nucleophosmin-negative tumors (P < 0.01, log-rank test).

Multivariate analysis done on nucleophosmin staining and clinical stage, identified as significant prognostic factors by univariate analysis, revealed that both significantly correlated, as separate variables, with overall survival (P = 0.0063; relative risk, 7.768; 95% confidence interval, 1.783-33.841 and P = 0.0039; relative risk, 5.964; 95% confidence interval, 1.773-20.060, respectively; Table 3).

On a second univariate analysis, we found that nucleophosmin positivity was also a strong negative predictor of overall survival in the 29 of 34 patients that had localized disease at diagnosis (P < 0.01; log-rank test; Fig. 4C).

The identification of novel prognostic biomarkers for Ewing's sarcoma is required to improve the management of Ewing's sarcoma. Global genomic and transcriptomic expression studies conducted to identify prognostic biomarkers for Ewing's sarcoma resulted in the identification of MTA1, CDH11 (14), STEAP1, NKX2-2, and CCND1 (15), gains in chromosomes 1q, 8, and 12 and deletions of 1p as genetic lesions implicated in the progression of Ewing's sarcoma (1618). Although these comprehensive studies may have the potential to further increase our understanding of the biology of Ewing's sarcoma and to lead to the development of practical tumor markers to support individualized therapy, practical prognostic biomarkers of Ewing's sarcoma are presently not used in a clinical setting.

Proteomic studies have unique advantages on other omics studies. The proteome is a functional translation of the genome, directly regulating cell phenotypes, and is thus a rich source of biomarkers. With this notion, we have established the gel-based proteomics system for cancer research and applied it to the Ewing's sarcoma proteomic study presented here. This is the first report using a proteomic approach to develop prognostic biomarkers for Ewing's sarcoma.

We identified 6 down-regulated and 47 up-regulated proteins in Ewing's sarcoma cases with poor prognosis. Functional classification revealed that these identified proteins belonged to a variety of functional pathways, including cytoskeletal/structural organization, transcription/translation, signal transduction, transport, antiapoptosis, response-oxidative stress, acute-phase response, and cell proliferation. The results of functional classification may suggest that the proteomic alterations observed may be a part of global series of functionally interconnected molecular lesions that include transcriptional and translational aberrations, which, taken together, include both the causes and the results of carcinogenesis and cancer progression.

This proteome study identified or confirmed the presence of several molecular aberrations concerning Ewing's sarcoma. The 47 proteins found to be up-regulated included neuron-specific enolase, which has been found to be associated with poor prognosis in Ewing's sarcoma (38, 39).

Nucleophosmin was included in the 47 proteins found to be up-regulated. Nucleophosmin overexpression has been related to carcinogenesis and tumor progression in prostate (25), gastric (26), colon (27), ovarian (28), and urinary bladder carcinomas (29). However, the association of nucleophosmin with Ewing's sarcoma has not been reported previously, including, importantly, in previous genomic and transcriptomic studies of Ewing's sarcoma (1419). This may be due to discordance between mRNA and protein expression, the fact that a different patient population was studied, or, finally, the fact that transcriptome and proteome studies cannot uncover entire genome data. These results, therefore, also suggest that studies using proteomic tools are able to reveal unique molecular aspects of Ewing's sarcoma.

Network analysis showed that nucleophosmin is linked with four proteins (c-myc, nuclear factor-κB, Sp1 and p53), which have also been found to be implicated in poor prognosis in Ewing's sarcoma (Supplementary Fig. S1). c-myc has been identified as a potential EWS-ets target gene (40) and as promoting malignant progression of Ewing's sarcoma (41). Activation of nuclear factor-κB was found to contribute to resistance of Ewing's sarcoma cells to apoptosis (42). Coexpression of Sp1 with EWS-ets oncoprotein enhances activation of vascular endothelial growth factor, the expression of which was shown to be a negative predictor of survival in Ewing's sarcoma (43). Aberrations in p53 were found in ∼10% of Ewing's sarcoma cases and were associated with shorter survival (44, 45). Taken together, these observations suggest that nucleophosmin can be a single biomarker probably by linking these functionally different proteins. Ewing's sarcoma is characterized by a translocation between the EWS gene and a member of the ETS family of transcriptional factors (46). The EWS/ETS fusion protein has an altered transcriptional activity and modulates the expression of several downstream target genes (47, 48). The association between the EWS/ETS fusion protein and nucleophosmin should be further investigated.

Nucleophosmin may be used as a novel prognostic biomarker of patients with Ewing's sarcoma. We found that nucleophosmin expression correlated with clinical outcome in 34 Ewing's sarcoma patients. Univariate and multivariate analyses revealed that nucleophosmin expression along with clinical stage (presence of metastases at diagnosis) was an independent prognostic factor in Ewing's sarcoma patients. Furthermore, nucleophosmin expression was also a significant prognostic factor in patients with localized disease. Applying these findings in a clinical setting poses the next challenge. As the incisional biopsy is a procedure done routinely in establishing the diagnosis in Ewing's sarcoma, the immunohistochemical examination of nucleophosmin expression can be done without any additional invasive examinations.

Although previous reports have suggested a possible association between nucleophosmin and malignancies, the functional role of nucleophosmin in Ewing's sarcoma is still unclear. Nucleophosmin overexpression has been reported to be involved in human tumorigenesis (49, 50). In one study, it led to increased proliferation and inhibition of apoptosis in tumor cells; overexpression of nucleophosmin reduced the percentage of cells in the G1 phase and increased the S-phase population in the p53-negative cells but induced cell cycle arrest in normal cells. Conducting further basic research on the function of nucleophosmin will pave the way for further understanding of the molecular background of Ewing's sarcoma and, hopefully, for novel diagnostic and therapeutic applications.

In conclusion, global protein expression profiling revealed the proteomic background of Ewing's sarcoma and identified novel associations of several proteins with progression of Ewing's sarcoma. Of the proteins with expression that may have prognostic value, we successfully validated the association of nucleophosmin expression with poor prognosis. The expression of the other proteins may still have prognostic value and further validation studies may prove it. Evaluation of nucleophosmin expression may allow the identification of poor prognosis Ewing's sarcoma patients who may benefit from highly effective treatment in the future.

No potential conflicts of interest were disclosed.

Grant support: Ministry of Health, Labor and Welfare and Program for Promotion of Fundamental Studies in Health Sciences of the National Institute of Biomedical Innovation of Japan.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

We thank Chizu Kina and Sachiko Miura for excellent technical support in the immunohistochemical study and Yukiko Fujie for excellent technical support in electrophoresis.

1
Cotterill SJ, Ahrens S, Paulussen M, et al. Prognostic factors in Ewing's tumor of bone: analysis of 975 patients from the European Intergroup Cooperative Ewing's Sarcoma Study Group.
J Clin Oncol
2000
;
18
:
3108
–14.
2
Atra A, Whelan JS, Calvagna V, et al. High-dose busulphan/melphalan with autologous stem cell rescue in Ewing's sarcoma.
Bone Marrow Transplant
1997
;
20
:
843
–6.
3
Diaz MA, Vicent MG, Madero L. High-dose busulfan/melphalan as conditioning for autologous PBPC transplantation in pediatric patients with solid tumors.
Bone Marrow Transplant
1999
;
24
:
1157
–9.
4
Bernstein ML, Devidas M, Lafreniere D, et al. Intensive therapy with growth factor support for patients with Ewing tumor metastatic at diagnosis: Pediatric Oncology Group/Children's Cancer Group Phase II Study 9457—a report from the Children's Oncology Group.
J Clin Oncol
2006
;
24
:
152
–9.
5
Engelhardt M, Zeiser R, Ihorst G, Finke J, Muller CI. High-dose chemotherapy and autologous peripheral blood stem cell transplantation in adult patients with high-risk or advanced Ewing and soft tissue sarcoma.
J Cancer Res Clin Oncol
2007
;
133
:
1
–11.
6
McTiernan A, Driver D, Michelagnoli MP, Kilby AM, Whelan JS. High dose chemotherapy with bone marrow or peripheral stem cell rescue is an effective treatment option for patients with relapsed or progressive Ewing's sarcoma family of tumours.
Ann Oncol
2006
;
17
:
1301
–5.
7
Grier HE, Krailo MD, Tarbell NJ, et al. Addition of ifosfamide and etoposide to standard chemotherapy for Ewing's sarcoma and primitive neuroectodermal tumor of bone.
N Engl J Med
2003
;
348
:
694
–701.
8
Kolb EA, Kushner BH, Gorlick R, et al. Long-term event-free survival after intensive chemotherapy for Ewing's family of tumors in children and young adults.
J Clin Oncol
2003
;
21
:
3423
–30.
9
Paulussen M, Ahrens S, Burdach S, et al. Primary metastatic (stage IV) Ewing tumor: survival analysis of 171 patients from the EICESS studies. European Intergroup Cooperative Ewing Sarcoma Studies.
Ann Oncol
1998
;
9
:
275
–81.
10
Bacci G, Ferrari S, Bertoni F, et al. Prognostic factors in nonmetastatic Ewing's sarcoma of bone treated with adjuvant chemotherapy: analysis of 359 patients at the Istituto Ortopedico Rizzoli.
J Clin Oncol
2000
;
18
:
4
–11.
11
Rodriguez-Galindo C, Spunt SL, Pappo AS. Treatment of Ewing sarcoma family of tumors: current status and outlook for the future.
Med Pediatr Oncol
2003
;
40
:
276
–87.
12
Hayes FA, Thompson EI, Meyer WH, et al. Therapy for localized Ewing's sarcoma of bone.
J Clin Oncol
1989
;
7
:
208
–13.
13
Marina NM, Pappo AS, Parham DM, et al. Chemotherapy dose-intensification for pediatric patients with Ewing's family of tumors and desmoplastic small round-cell tumors: a feasibility study at St. Jude Children's Research Hospital.
J Clin Oncol
1999
;
17
:
180
–90.
14
Ohali A, Avigad S, Zaizov R, et al. Prediction of high risk Ewing's sarcoma by gene expression profiling.
Oncogene
2004
;
23
:
8997
–9006.
15
Cheung IY, Feng Y, Danis K, et al. Novel markers of subclinical disease for Ewing family tumors from gene expression profiling.
Clin Cancer Res
2007
;
13
:
6978
–83.
16
Armengol G, Tarkkanen M, Virolainen M, et al. Recurrent gains of 1q, 8 and 12 in the Ewing family of tumours by comparative genomic hybridization.
Br J Cancer
1997
;
75
:
1403
–9.
17
Maurici D, Perez-Atayde A, Grier HE, Baldini N, Serra M, Fletcher JA. Frequency and implications of chromosome 8 and 12 gains in Ewing sarcoma.
Cancer Genet Cytogenet
1998
;
100
:
106
–10.
18
Hattinger CM, Potschger U, Tarkkanen M, et al. Prognostic impact of chromosomal aberrations in Ewing tumours.
Br J Cancer
2002
;
86
:
1763
–9.
19
Schaefer KL, Eisenacher M, Braun Y, et al. Microarray analysis of Ewing's sarcoma family of tumours reveals characteristic gene expression signatures associated with metastasis and resistance to chemotherapy.
Eur J Cancer
2008
;
44
:
699
–709.
20
Petricoin EF, Ardekani AM, Hitt BA, et al. Use of proteomic patterns in serum to identify ovarian cancer.
Lancet
2002
;
359
:
572
–7.
21
Suehara Y, Kondo T, Fujii K, et al. Proteomic signatures corresponding to histological classification and grading of soft-tissue sarcomas.
Proteomics
2006
;
6
:
4402
–9.
22
Chen G, Gharib TG, Wang H, et al. Protein profiles associated with survival in lung adenocarcinoma.
Proc Natl Acad Sci U S A
2003
;
100
:
13537
–42.
23
Suehara Y, Kondo T, Seki K, et al. Pfetin as a prognostic biomarker of gastrointestinal stromal tumors revealed by proteomics.
Clin Cancer Res
2008
;
14
:
1707
–17.
24
Okano T, Kondo T, Fujii K, et al. Proteomic signature corresponding to the response to gefitinib (Iressa, ZD1839), an epidermal growth factor receptor tyrosine kinase inhibitor in lung adenocarcinoma.
Clin Cancer Res
2007
;
13
:
799
–805.
25
Subong EN, Shue MJ, Epstein JI, Briggman JV, Chan PK, Partin AW. Monoclonal antibody to prostate cancer nuclear matrix protein (PRO:4-216) recognizes nucleophosmin/B23.
Prostate
1999
;
39
:
298
–304.
26
Tanaka M, Sasaki H, Kino I, Sugimura T, Terada M. Genes preferentially expressed in embryo stomach are predominantly expressed in gastric cancer.
Cancer Res
1992
;
52
:
3372
–7.
27
Nozawa Y, Van Belzen N, Van der Made AC, Dinjens WN, Bosman FT. Expression of nucleophosmin/B23 in normal and neoplastic colorectal mucosa.
J Pathol
1996
;
178
:
48
–52.
28
Zhang Y. The ARF-B23 connection: implications for growth control and cancer treatment.
Cell Cycle
2004
;
3
:
259
–62.
29
Tsui KH, Cheng AJ, Chang PL, Pan TL, Yung BY. Association of nucleophosmin/B23 mRNA expression with clinical outcome in patients with bladder carcinoma.
Urology
2004
;
64
:
839
–44.
30
Wolf RE, Enneking WF. The staging and surgery of musculoskeletal neoplasms.
Orthop Clin North Am
1996
;
27
:
473
–81.
31
Shankar AG, Ashley S, Craft AW, Pinkerton CR. Outcome after relapse in an unselected cohort of children and adolescents with Ewing sarcoma.
Med Pediatr Oncol
2003
;
40
:
141
–7.
32
Urano F, Umezawa A, Yabe H, et al. Molecular analysis of Ewing's sarcoma: another fusion gene, EWS-E1AF, available for diagnosis.
Cancer Sci
1998
;
89
:
703
–11.
33
Kondo T, Hirohashi S. Application of highly sensitive fluorescent dyes (CyDye DIGE Fluor saturation dyes) to laser microdissection and two-dimensional difference gel electrophoresis (2D-DIGE) for cancer proteomics.
Nat Protoc
2006
;
1
:
2940
–56.
34
Kaplan EL, Meier P. Nonparametric estimation from incomplete observations.
J Am Stat Assoc
1958
;
53
:
1
.
35
Cox DR. Regression models and life tables.
J R Stat Soc
1972
;
34
:
187
–220.
36
Obata H, Ueda T, Kawai A, et al. Clinical outcome of patients with Ewing sarcoma family of tumors of bone in Japan: the Japanese Musculoskeletal Oncology Group cooperative study.
Cancer
2007
;
109
:
767
–75.
37
Yun JP, Miao J, Chen GG, et al. Increased expression of nucleophosmin/B23 in hepatocellular carcinoma and correlation with clinicopathological parameters.
Br J Cancer
2007
;
96
:
477
–84.
38
Bacci G, Ferrari S, Bertoni F, et al. Neoadjuvant chemotherapy for peripheral malignant neuroectodermal tumor of bone: recent experience at the Istituto Rizzoli.
J Clin Oncol
2000
;
18
:
885
–92.
39
Oberlin O, Deley MC, Bui BN, et al. Prognostic factors in localized Ewing's tumours and peripheral neuroectodermal tumours: the third study of the French Society of Paediatric Oncology (EW88 study).
Br J Cancer
2001
;
85
:
1646
–54.
40
Bailly RA, Bosselut R, Zucman J, et al. DNA-binding and transcriptional activation properties of the EWS-FLI-1 fusion protein resulting from the t(11;22) translocation in Ewing sarcoma.
Mol Cell Biol
1994
;
14
:
3230
–41.
41
Sollazzo MR, Benassi MS, Magagnoli G, et al. Increased c-myc oncogene expression in Ewing's sarcoma: correlation with Ki67 proliferation index.
Tumori
1999
;
85
:
167
–73.
42
Javelaud D, Poupon MF, Wietzerbin J, Besancon F. Inhibition of constitutive NF-κB activity suppresses tumorigenicity of Ewing sarcoma EW7 cells.
Int J Cancer
2002
;
98
:
193
–8.
43
Fuchs B, Inwards CY, Janknecht R. Vascular endothelial growth factor expression is up-regulated by EWS-ETS oncoproteins and Sp1 and may represent an independent predictor of survival in Ewing's sarcoma.
Clin Cancer Res
2004
;
10
:
1344
–53.
44
Abudu A, Mangham DC, Reynolds GM, et al. Overexpression of p53 protein in primary Ewing's sarcoma of bone: relationship to tumour stage, response and prognosis.
Br J Cancer
1999
;
79
:
1185
–9.
45
de Alava E, Antonescu CR, Panizo A, et al. Prognostic impact of P53 status in Ewing sarcoma.
Cancer
2000
;
89
:
783
–92.
46
Delattre O, Zucman J, Plougastel B, et al. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours.
Nature
1992
;
359
:
162
–5.
47
Ladanyi M. EWS-FLI1 and Ewing's sarcoma: recent molecular data and new insights.
Cancer Biol Ther
2002
;
1
:
330
–6.
48
Siligan C, Ban J, Bachmaier R, et al. EWS-FLI1 target genes recovered from Ewing's sarcoma chromatin.
Oncogene
2005
;
24
:
2512
–24.
49
Itahana K, Bhat KP, Jin A, et al. Tumor suppressor ARF degrades B23, a nucleolar protein involved in ribosome biogenesis and cell proliferation.
Mol Cell
2003
;
12
:
1151
–64.
50
Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer.
Nat Rev Cancer
2006
;
6
:
493
–505.