Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2010 Feb 1;28(7):1240–1246. doi: 10.1200/JCO.2008.21.1268

Gene Expression Profiling for Survival Prediction in Pediatric Rhabdomyosarcomas: A Report From the Children's Oncology Group

Elai Davicioni 1, James R Anderson 1, Jonathan D Buckley 1, William H Meyer 1, Timothy J Triche 1,
PMCID: PMC3040045  PMID: 20124188

Abstract

Purpose

We investigated whether tumors from diagnostic biopsies of primary rhabdomyosarcoma (RMS) contain relevant prognostic information in the form of gene expression signatures that can be used to model and predict outcome of patients.

Patients and Methods

A 22,000-probe set microarray was used to evaluate 120 RMS specimens and correlate gene expression patterns to survival. Multivariate gene expression models or metagenes were developed using cross-validated Cox regression proportional hazards modeling and were evaluated using Kaplan-Meier analysis.

Results

A 34-metagene, based on expression patterns of 34 genes, was highly predictive of outcome. It was not highly correlated with individual clinical risk factors such as patient age, stage, tumor size, or histology. However, it was correlated with a risk classification used by the Children's Oncology Group and the biologic subsets of alveolar histology tumors.

Conclusion

These data support further evaluation of RMS metagenes to discriminate patients with good prognosis from those with poor prognosis, with the potential to direct risk-adapted therapy.

INTRODUCTION

The cure rate for patients with rhabdomyosarcoma (RMS) is more than 70% for patients with nonmetastatic disease,1 and much of this realized gain over the past few decades can be attributed to the use of intensive multimodal therapy including surgery, radiotherapy, and chemotherapy. This cure rate is not expected to change significantly until targeted tumor-specific agents are developed. Because multimodal therapy can be associated with acute toxicities and long-term adverse effects, such as growth and developmental defects, one of the major areas for improvement relates to quality of life for young cancer survivors.2 Recent studies have revealed that some patients can be treated effectively without radiotherapy3 and with less intensive chemotherapy,4 reducing acute and long-term adverse effects.5 Moreover, there appear to be subsets of patients with metastatic disease at diagnosis with an atypically more favorable outcome.6,7

A crucial determinant of the overall success of such risk-adapted therapy is the effectiveness of clinical staging systems for patient prognosis and treatment assignment. Various forms of clinicopathologic staging have been used to define risk in several international clinical trial groups over the past several decades, and the latest development used in ongoing clinical trials of the Children's Oncology Group (COG; under the auspices of the COG Soft Tissue Sarcoma Committee) is a three-tier risk classification system. The COG risk classification system incorporates both of the earlier postsurgical clinical group and TNM stage schemes as well as tumor histology,8 and it appears to be the most powerful prognostic scheme devised to date.9

One issue with the COG risk classification system is that many patients fall into the intermediate-risk category where survival is most heterogeneous,10 suggesting that even the best clinical risk model has difficulty in identifying some aspects of underlying biology of tumors, in particular relating to their clinical aggressiveness.11 Molecular staging using, for example, gene expression profiles has promise in predicting long-term patient outcome by analysis of the tumor at diagnosis.12 An inherent assumption of this approach, supported by recent analyses13,14 is the hypothesis that every tumor contains informative gene expression signatures that, at the time of diagnosis, can predict the biologic behavior of the tumor over time.15 A powerful approach for modeling patient survival data is using Cox regression proportional hazards models; recently, this has been applied to gene expression data sets11,1618 in efforts to generate true continuous predictors of survival that are independent of clinicopathologic variables in predicting treatment outcome.11 In this proof-of-concept study, we describe the development of a metagene or multivariable continuous predictor of outcome using Cox regression–based modeling for 120 patients with RMS.

PATIENTS AND METHODS

Tumor Specimens

Tumor specimens used to develop outcome prediction models were, as recently reported,19 obtained from the Intergroup Rhabdomyosarcoma Study Group (IRSG)/Pediatric Cooperative Human Tissue Network (Columbus, OH) and Childrens Hospital Los Angeles (CHLA) institutional tumor banks from 120 patients who were enrolled in IRSG IV and V COG clinical trials. Clinical covariates were obtained from the COG Statistics and Data Center (Arcadia, CA). Two patients had mixed alveolar/embryonal histology and were considered alveolar for the purposes of this analysis. From the previously reported study, we selected only those patients with RMS histology (alveolar, embryonal, spindle-cell, and botryoid) on review diagnosis (ie, excluded all patients with non-RMS soft tissue sarcoma or undifferentiated sarcoma) and those with sufficient follow-up data (ie, alive [censored] patients with < 2 years of follow-up were omitted) for this analysis. Sample preparation and Affymetrix GeneChip Human U133A (Affymetrix, Santa Clara, CA) microarray protocols were previously described.19,20 Complete microarray protocols can be found at the University of Southern California (USC)/CHLA Genome Core Web site at http://genomecore-chla.usc.edu/GenomeCore/GenomeCore.html.

Analysis of Gene Expression

All data management and analysis was conducted using the Genetrix suite of tools for microarray analysis (Epicenter Software, http://www.epicentersoftware.com). Probe set modeling and data preprocessing were derived using the robust multi-array algorithm implemented within the ProbeProfiler module (Corimbia, Berkeley, CA). The full data set of 22,215 probe sets was reduced to 21,718 probe sets (henceforth, genes) by eliminating genes with a standard deviation of less than 10 Affymetrix difference intensity units of a normalized data range, and the data were log transformed. The complete tumor microarray data set (including sample covariate data) can be found on the National Cancer Institute (NCI) Cancer Array Database at https://array.nci.nih.gov/caarray/project/trich-00099.

Metagene Construction and Evaluation

Metagenes were constructed as previously described,20 using Cox proportional hazards modeling of RMS gene expression data under cross-validation. Genes were ranked and selected using sampling statistics obtained across multiple iterations testing for significance in both training (n = 60) and testing (n = 60) randomized subsets. Weighting factors were obtained from the signed square root of the Cox χ2 test statistic modeled on the entire cohort. The metagene score for each patient was calculated as a weighted sum of the gene expression value. Detailed descriptions of the data analysis can be found in the Data Supplement (online only).

Survival Analysis

Comparison of survival times was carried out using Kaplan-Meier survival plots and log-rank tests of significance. Comparisons between molecular groups and tests of association used Fisher's exact or χ2 tests to compare the frequency distributions of patient characteristics. Multivariate tests for association of factors with survival used a Cox regression proportional hazards model.

RESULTS

Generation of Multigene Prognostic Models

A cohort of 120 pediatric RMS patient tumor samples (Supplementary Table 1, online only) with at least 2 years censored follow-up data after diagnosis (except patients who died of disease at any time point) were used to identify genes correlated to overall survival (OS) times with Cox proportional hazards regression modeling. Most of the deaths (67%) occurred within the first 2 years of diagnosis and the cause of death was attributed to the tumor in all patients except for two (one from infection on regimen and one from toxicity unrelated to the chemotherapy regimen). Of the patients who died, 24 (62%) had alveolar histology, 13 (33%) had embryonal histology, and two (5%) had mixed alveolar/embryonal histology. No reported deaths occurred in patients with spindle-cell or botryoid histology tumors.

Using Cox modeling of OS with log-transformed gene expression data (see Patients and Methods), 578 genes with significant Cox χ2 scores over 2,500 iterations of the algorithm (P < .01) were identified (Supplementary Table 2, online only). Next, multigene continuous predictors of outcome were assembled and evaluated as described previously20 (see schematic in Appendix Fig A1, online only). The maximum likelihood estimate of the χ2 test statistics were determined for each multivariate model and showed that a 34-probe set model or 34-metagene (MG34; Table 1) had the highest significance score (blue curve, Appendix Fig A1). By permuting the gene expression data and generating new metagenes from the permuted data set, we show that permuted models do not reach statistical significance, indicating that these results are not likely due to chance alone (red curve, Appendix Fig A2, online only).

Table 1.

Genes Used to Create the 34-Metagene Continuous Predictor of Outcome

Affymetrix ID Gene Name Gene Symbol
Genes correlated to good patient outcome
    214643_x_at Bridging integrator 1 BIN1
    219953_s_at Chromosome 11 open reading frame 17 C11orf17
    218314_s_at Chromosome 11 open reading frame 57 C11orf57
    201905_s_at CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase-like CTDSPL
    204643_s_at Ecto-NOX disulfide-thiol exchanger 2 ENOX2
    218695_at Exosome component 4 EXOSC4
    207688_s_at Inhibin, beta C INHBC
    222250_s_at Integrator complex subunit 7 INTS7
    202788_at Mitogen-activated protein kinase-activated protein kinase 3 MAPKAPK3
    213946_s_at Obscurin-like 1 OBSL1
    35156_at R3H domain and coiled-coil containing 1 R3HCC1
    218392_x_at Sideroflexin 1 SFXN1
    207069_s_at SMAD, mothers against DPP homolog 6 (Drosophila) SMAD6
    214662_at WD repeat domain 43 WDR43
    219548_at Zinc finger protein 16 (KOX 9) ZNF16
Genes correlated to poor patient outcome
    221588_x_at Aldehyde dehydrogenase 6 family, member A1 ALDH6A1
    211248_s_at Chordin CHRD
    210656_at Embryonic ectoderm development EED
    212546_s_at FRY-like 1 FRYL
    209525_at Hepatoma-derived growth factor, related protein 3 HDGFRP3
    220447_at Histamine receptor H3 HRH3
    209184_s_at Insulin receptor substrate 2 IRS2
    204075_s_at KIAA0562 KIAA0562
    204584_at L1 cell adhesion molecule L1CAM
    213672_at Methionine-tRNA synthetase MARS
    215921_at Nuclear pore complex interacting protein-like 1 NPIPL1
    209791_at Peptidyl arginine deiminase, type II PADI2
    205632_s_at Phosphatidylinositol-4-phosphate 5-kinase, type I, beta PIP5K1B
    211974_x_at Recombination signal binding protein for immunoglobulin kappa J region RBPJ
    218394_at Rogdi homolog (Drosophila) ROGDI
    213437_at RUN and FYVE domain-containing 2; Run- and FYVE-domain containing protein RUFY3
    219196_at Secretogranin III SCG3
    213434_at Syntaxin 2 STX2
    202342_s_at Tripartite motif-containing 2 TRIM2

Post Hoc Analysis of MG34

To validate the performance of the MG34, RMS patients were split into three groups (tertiles, determined by the histogram bar groupings) by their computed metagene predictor scores (Fig 1A). The mean metagene predictor scores for the third tertile were five- and 17-fold greater than second and first tertile patient scores, respectively. Kaplan-Meier analysis revealed that patients in the first (n = 39; blue curve) and second (n = 41; green curve) tertiles had 5-year OS rates of 98% and 75%, respectively (Fig 1B). In contrast, patients in the third tertile (n = 40; red curve) had a 5-year OS rate of only 29% and median survival of 24 months. Of the cohort of 120 patients, there were seven patients for whom missing stage or clinical group data meant that we could not classify them into the risk groups now used by the COG (in current clinical trials for RMS). The remaining 113 patients were grouped according to the COG risk groups as indicated in Table 2. Kaplan-Meier analysis (Fig 1C) shows that the 5-year OS rates for patients in this cohort grouped according to the COG risk category criteria are reflective of the survival rates observed in larger cohorts from COG clinical trials.10 We also observed that MG34 tertile groups are highly correlated to COG risk groups; for example, most high-risk patients are found in the third tertile, whereas few clinically low-risk patients are within this tertile (Table 2).

Fig 1.

Fig 1.

Metagene predictor scores determine outcome in rhabdomyosarcoma patients. (A) Histogram showing the binned distribution of the 34-metagene predictor scores for 120 patients (vertical purple lines highlight the tertile cut points). Kaplan-Meier survival analysis of all 120 rhabdomyosarcoma patients (B) using tertiles as groups and (C) for 113 RMS patients with known Children's Oncology Group (COG) risk groups (Table 2). Numbers below the curves indicate the number of patients at risk, and P values are from log-rank test. Int, intermediate.

Table 2.

Comparison of COG Risk Groups and Metagene Tertile Groups

COG Risk Group Risk Group Criteria 5-Year OS, (%)* No. of Patients in MG34 Tertiles
1st 2nd 3rd
Low Embryonal histology and stage 1 or stage 2/3, group I/II 90 19 11 2
Intermediate Embryonal histology and stage 2/3, group III or alveolar histology, groups I-III 77 12 22 14
High All patients with group IV disease 24 5 7 21
5-Year OS (%)*
95 76 29

Abbreviations: COG, Children's Oncology Group; OS, overall survival; MG34, 34-metagene, based on expression patterns of 34 genes.

*

Log-rank test; P < .001.

Next, we looked at the predictive value of the MG34 tertiles within the COG risk groups. COG low-risk patients (n = 32) were mostly in the MG34 first tertile (n = 19) or second tertile (n = 11) except for two embryonal histology (stage 3, group II disease) patients who were categorized in the third tertile (log-rank P < .013). While the log-rank test for the comparison of survival is < 0.05, it is mostly the result of early failure of one of these patients in the third tertile (Fig 2A). For intermediate-risk patients (n = 48), there appears to be clear evidence that the MG34 tertiles are predictive of survival (Fig 2B). For COG intermediate-risk group patients in the MG34 first tertile, the 5-year OS rate was 100% (n = 12), whereas second tertile (n = 22) and third tertile (n = 14) patients had 5-year OS rates of 86% and 43%, respectively (log-rank P < .00003). Of note, we observed that 71% (12 of 17) of COG intermediate-risk patients with tumors expressing the PAX3-FKHR fusion gene were in the MG34 third tertile, the one that is most different in terms of survival. In contrast, six of seven PAX7-FKHR and three of three fusion-negative alveolar histology tumors from the COG intermediate-risk group were in the MG34 second tertile (Supplementary Table 3, online only). Therefore, it appears that for intermediate-risk patients, higher MG34 scores (eg, third tertile) are tightly correlated to the PAX3-FKHR alveolar subtype (Supplementary Table 4, online only). For patients in the COG high-risk group, 64% (21 of 33) were in the MG34 third tertile. Five patients with group IV disease (four embryonal, one alveolar) and improved survival were categorized into the first tertile, but for the remainder of these patients, there was no appreciable difference in the survival curves between the second MG34 tertile (n = 7) and the third MG34 tertile (n = 21), except median survival was 33 months versus 22 months, respectively (Fig 2C). Comparison of the metagene predictor scores with clinical risk factors can be found in Supplementary Table 5 (online only). Appendix Figure A3 (online only) shows the distribution of MG34 scores within histologic and genetic subtypes.

Fig 2.

Fig 2.

Evaluation of metagene predictor scores within Children's Oncology Group (COG) risk groups. Kaplan-Meier survival analysis of COG low-risk (A), intermediate-risk (B), and high-risk (C) patients using 34-metagene tertiles as groups. Numbers below the curves indicate the number of patients at risk, and P values are from log-rank test.

DISCUSSION

Previous gene expression profiling of RMS patient tumors by our group and by others1922 focused primarily on resolving issues of diagnosis and enhancing the understanding of tumor classification from a genome-wide perspective. While a 2006 study20 showed that expression signatures of putative PAX-FKHR target genes may be of prognostic value in the subset of PAX-FKHR translocation-positive alveolar RMS patients and a 2009 report19 showed differences in prognosis for molecular-based classes of RMS tumors, these findings have not yet had an impact on clinical practice for patient stratification or assignment to treatment protocols. The main reason is that they do not seem to add much more prognostic information beyond that captured by established pathologic criteria, such as favorable (ie, embryonal) versus unfavorable (ie, alveolar) tumor histology, known for nearly three decades as an independent prognostic factor. The MG34 described here appears to discriminate patient risk independent of tumor histology and, as a continuous rather than discrete variable, it reflects the spectrum of differential gene expression observed in this heterogeneous group of tumors.

Genes such as L1CAM that are highly expressed in poor-outcome patients, a cell adhesion molecule,2325 and IRS226 are associated with increased metastatic potential and invasiveness in several tumor types. Another poor-outcome gene, transcription factor RBPSJ, is involved in repression of differentiation in numerous cancers.27 Conversely, a good-outcome gene, BIN1, is a well-characterized tumor suppressor gene that promotes muscle differentiation28 and differentiation of tumor cells.29 Though the functional relationship of MG34 genes in determining tumor behavior and hence outcome of RMS patients is at present unclear, these and many others (Supplementary Table 2) appear to impart independent prognostic information. In addition, we have shown that the MG34 model is one of numerous expression signatures correlated to patient prognosis (Appendix Fig A2), as has been demonstrated in other tumor systems.30,31

We previously reported a PAX-FKHR 33-metagene that predicted outcome in a subset of alveolar RMS patients whose tumors expressed products of PAX-FKHR fusion genes. This PAX-FKHR 33-metagene and the present pan-RMS MG34 do not show any overlap. This is not surprising given the fact that the PAX-FKHR metagene was generated from a list of putative PAX-FKHR targets derived from expression analysis of an in vitro model system (ectopic expression of PAX-FKHR in ERMS RD cell lines) and PAX-FKHR–positive primary tumors only. However, five genes (MYLPF, TNNC2, IL4R, NELL1, and BMP5) from the cell line model system analysis were also identified in the present analysis of outcome-correlated genes in all RMS tumors (Supplementary Table 2) though they were not incorporated into the MG34 model reported here. Our working hypothesis is that PAX3 and PAX7 (and their cognate PAX-FKHR fusion genes in alveolar RMS) activate a unique transcriptional program that confers rhabdomyosarcoma-ness in general (eg, myogenic phenotype in a sarcoma). Furthermore, the PAX-FKHR fusion proteins in alveolar RMS are believed to further activate a transcriptional program that confers a more aggressive phenotype (perhaps in part characterized by some of the genes identified here and in the 2006 study). Previous work from the Barr group32 demonstrated that levels of PAX-FKHR are also crucial where PAX-FKHR overexpressed in cell lines caused transformation at lower levels and growth suppression at higher levels. Multiple functionally significant splice forms of PAX-FKHR may have implications for tumor phenotypes such as clinical aggressiveness and the correlation between fusion gene expression; wild-type PAX3/7 expression and other factors yet to be identified likely have important roles in conferring different biologic properties to RMS cells.3235 In a follow-on study now underway on a larger cohort of RMS patients with higher resolution exon microarrays, we intend to address the question of whether RMS patient survival can be better modeled with separate metagenes for PAX-FKHR and fusion-negative RMS.

Kaplan-Meier analysis of the objectively derived MG34 tertiles shows patients divided into three highly disparate groups in terms of survival, which suggests that MG34 tertiles are predictive of survival. Additionally, MG34 tertiles are correlated with the COG clinicopathologic risk category used to assign patients with RMS to treatment studies. This is perhaps not surprising because both depend on features related to the biology of the tumors, although they are measured in different ways. All but two of the patients who were low risk by clinicopathologic features had tumors classified into the first or second MG34 tertiles. Most had embryonal disease, and survival did not appear to differ significantly by whether low-risk patients were in the first or second tertile. Most patients (21 of 33) who are high risk by clinicopathologic features had tumors classified into the third MG34 tertile. Patients in the COG intermediate-risk group had tumors evenly distributed across the MG34 tertiles. Poorer survival outcomes were observed for third-tertile patients within the COG intermediate-risk group, with the majority of these patients (12 of 14) having PAX3-FKHR and alveolar histology disease. The poorest prognosis MG34 third tertile appears to be associated with PAX3-FKHR alveolar histology disease; overall, 63% (25 of 40) of the MG34 third-tertile patients have PAX3-FKHR alveolar tumors, whereas the percentages are only 3% (one of 39) and 10% (four of 41) for the first and second tertiles, respectively. In contrast, 73% (eight of 11) of PAX7-FKHR alveolar histology tumors were in the second tertile, supporting previous studies that report a more favorable outcome for this subset of alveolar patients.33,36

While the third tertile predominately comprised the less favorable PAX3-FKHR alveolar RMS tumors, it also included eight patients with embryonal disease and two with mixed alveolar/embryonal disease. Intriguingly, the metagene tertile risk groups varied most markedly in patients who presented with metastatic disease (group IV), which is the most adverse prognostic factor for RMS patients.6 While most of the metastatic disease patients were found in the third tertile group, nearly a third were found in the other two tertiles and, in accordance with previous observations, these (8 of 11) had primarily group IV embryonal histology tumors.6,37 These data suggest that a genomic-based classifier such as MG34 could be used to discern patients with high-risk disease who are most responsive to current therapeutic modalities and may provide a means to separate them from high-risk patients unlikely to respond to conventional chemotherapy regimens. This approach could therefore enable clinicians to better test experimental therapeutic agents on chemotherapy-naïve high-risk patients9 and make testing clinical trials for these agents more efficient.

Current methods for RMS staging have evolved to direct risk-adapted therapy9 using complex clinical risk models.38 This is important not only for patient management but also for evaluation of the effects of different treatment regimens in clinical trials. However, it appears that clinicopathologic-based staging systems do not identify many of the fundamental differences in underlying tumor biology.11 The MG34, a continuous predictor of patient outcome when split into tertile groups, performed similarly to the COG risk groups in a training cohort. This is notable since the MG34 risk groups were derived from statistical cut points (ie, tertile groups) and were not optimized by post hoc analysis. Further work is required to expand genome-wide analyses to further training and independent validation cohorts, efforts that will likely require hundreds of patient samples as has been done previously for analysis of other prognostic factors on routine clinical material (eg, formalin-fixed paraffin embedded tumor sections).7,22 The present work suggests that the focus should be on the intermediate-risk patients, the most prevalent type of RMS clinical trial patients. In this subgroup, the MG34 model appears to add prognostic information and separates out significant numbers of patients with more favorable or worse prognosis than the OS trends in this heterogeneous risk category. On the basis of these initial results, genomic classifiers for prognosis and the substratification of patients at the time of diagnosis have great promise as clinical tools to better the treatment, management, and outcomes for RMS patients.

Supplementary Material

Data Supplement
Publisher's Note

Acknowledgment

We would like to acknowledge our late colleague and friend James W. Jacobson, PhD, who passed on December 23, 2009 and send our deepest condolences to the Jacobson family. Jim was the chief of the Diagnostic Biomarkers and Technology Branch of the National Cancer Institute and was critical to ensuring that NCI funding be directed toward innovation in cancer diagnosis.

Appendix

Methods.

A Cox proportional hazards model was used to determine which genes should be included in the gene expression–based outcome predictor model (Appendix Fig A1). Multivariate Cox modeling was fitted with coefficients for each gene that best correlated with censored overall survival (OS) data. Positive coefficients were assigned to genes whose high expression was correlated to a low likelihood of survival, whereas negative coefficients were assigned to genes whose high expression was correlated to a high likelihood of survival. The metagene score for each patient was calculated as a weighted sum of the gene expression value, with the weights being the signed square root of the Cox χ2 test statistic. The multivariate model was developed by first identifying the best single-gene predictors of outcome (P < .05) by sample cross-validation. Leave-n-out cross-validation was used by randomly excluding 60 samples (ie, 50% of tumors) for each iteration of the model, running the Cox regression proportional hazards model on this training set. The remaining samples in each iteration, the test set, were then used to evaluate the results from running the model on the training set. This process was reiterated to generate 2,500 cross-validated models to calculate the number of times each gene was used in a cross-validated model, and genes were ranked by the number of models in which they were selected (ie, sampling statistics). Metagenes, or multigene (ie, multivariate) models were built in a step-wise procedure from the ranked list of the best single-gene predictors (determined by frequency of gene usage tabulated in the sampling statistics). Cox regression χ2 test statistics were determined for each multivariate model and showed that a 34-probe set (34-metagene) model had the highest χ2 test statistic. Next, the data set was permuted by sample shuffling and the cross-validated Cox regression modeling was repeated, generating metagenes from the ranked list of the best single-gene predictors in the permuted data set. The Cox regression χ2 statistics from the metagenes generated on the permuted data set indicate that these results are not likely due to chance alone (Appendix Fig A2).

Fig A1.

Fig A1.

Development, evaluation, and validation of metagene continuous predictors of rhabdomyosarcoma (RMS) patient outcome. Metagene models were developed using a model-building cohort of 120 patients from North American Intergroup Rhabdomyosarcoma Study (IRS) and Children's Oncology Group (COG) clinical trials under reiterative Cox regression modeling with 50% sample cross-validation.

Fig A2.

Fig A2.

Evaluation of the best metagene model. Metagenes were generated by building in a step-wise procedure from the ranked list of the best single gene predictors as determined by reiterative cross-validation. Cox regression χ2 values plotted for each metagene generated (blue curve, triangles) were compared with those generated from a permuted data set (gold curve) for the rhabdomyosarcoma metagenes built in a step-wise procedure from the ranked gene list (Supplementary Table 1). Arrow indicates the peak χ2 statistics for a 34-probe set metagene or 34-metagene. Generation of metagenes on a permuted data set indicates that the Cox regression model χ2 values are not likely attributed to chance alone (gold curve, circles).

Fig A3.

Fig A3.

Interquartile range of 34-metagene scores for postsurgical risk groups (A) and histopathologic subtypes (B). The box plots show the 25th and 75th percentiles, the error bars show the minimum and maximum scores, and the middle bars show the median score on a logarithmic scale. RMS, rhabdomyosarcoma.

Footnotes

Supported in part by Grants No. U01-CA-114757 from the Strategic Partnering to Evaluate Cancer Signatures (SPECS) program (T.J.T.) and No. U10 CA98543 from the Children's Oncology Group Chair, National Cancer Institute, National Institutes of Health, Bethesda, MD.

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO's conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors.

Employment or Leadership Position: Elai Davicioni, GenomeDx Biosciences (C); Jonathan D. Buckley, Epicenter Software (C) Consultant or Advisory Role: None Stock Ownership: Elai Davicioni, GenomeDx Biosciences; Jonathan D. Buckley, Epicenter Software Honoraria: None Research Funding: None Expert Testimony: None Other Remuneration: None

AUTHOR CONTRIBUTIONS

Conception and design: Elai Davicioni, Jonathan D. Buckley, Timothy J. Triche

Financial support: Timothy J. Triche

Administrative support: William H. Meyer, Timothy J. Triche

Provision of study materials or patients: James R. Anderson, William H. Meyer

Collection and assembly of data: Elai Davicioni, James R. Anderson, Timothy J. Triche

Data analysis and interpretation: Elai Davicioni, James R. Anderson, Jonathan D. Buckley, Timothy J. Triche

Manuscript writing: Elai Davicioni, James R. Anderson, William H. Meyer, Timothy J. Triche

Final approval of manuscript: Elai Davicioni, James R. Anderson, Jonathan D. Buckley, William H. Meyer, Timothy J. Triche

REFERENCES

  • 1.McDowell HP. Update on childhood rhabdomyosarcoma. Arch Dis Child. 2003;88:354–357. doi: 10.1136/adc.88.4.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Stevens MC. Treatment for childhood rhabdomyosarcoma: The cost of cure. Lancet Oncol. 2005;6:77–84. doi: 10.1016/S1470-2045(05)01733-X. [DOI] [PubMed] [Google Scholar]
  • 3.Schuck A, Mattke AC, Schmidt B, et al. Group II rhabdomyosarcoma and rhabdomyosarcomalike tumors: Is radiotherapy necessary? J Clin Oncol. 2004;22:143–149. doi: 10.1200/JCO.2004.04.180. [DOI] [PubMed] [Google Scholar]
  • 4.Crist WM, Anderson JR, Meza JL, et al. Intergroup rhabdomyosarcoma study-IV: Results for patients with nonmetastatic disease. J Clin Oncol. 2001;19:3091–3102. doi: 10.1200/JCO.2001.19.12.3091. [DOI] [PubMed] [Google Scholar]
  • 5.Meza JL, Anderson J, Pappo AS, et al. Analysis of prognostic factors in patients with nonmetastatic rhabdomyosarcoma treated on intergroup rhabdomyosarcoma studies III and IV: The Children's Oncology Group. J Clin Oncol. 2006;24:3844–3851. doi: 10.1200/JCO.2005.05.3801. [DOI] [PubMed] [Google Scholar]
  • 6.Breneman JC, Lyden E, Pappo AS, et al. Prognostic factors and clinical outcomes in children and adolescents with metastatic rhabdomyosarcoma: A report from the Intergroup Rhabdomyosarcoma Study IV. J Clin Oncol. 2003;21:78–84. doi: 10.1200/JCO.2003.06.129. [DOI] [PubMed] [Google Scholar]
  • 7.Oberlin O, Rey A, Lyden E, et al. Prognostic factors in metastatic rhabdomyosarcomas: Results of a pooled analysis from United States and European cooperative groups. J Clin Oncol. 2008;26:2384–2389. doi: 10.1200/JCO.2007.14.7207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Raney RB, Anderson JR, Barr FG, et al. Rhabdomyosarcoma and undifferentiated sarcoma in the first two decades of life: A selective review of intergroup rhabdomyosarcoma study group experience and rationale for Intergroup Rhabdomyosarcoma Study V. J Pediatr Hematol Oncol. 2001;23:215–220. doi: 10.1097/00043426-200105000-00008. [DOI] [PubMed] [Google Scholar]
  • 9.Breitfeld PP, Meyer WH. Rhabdomyosarcoma: New windows of opportunity. Oncologist. 2005;10:518–527. doi: 10.1634/theoncologist.10-7-518. [DOI] [PubMed] [Google Scholar]
  • 10.Meyer WH, Spunt SL. Soft tissue sarcomas of childhood. Cancer Treat Rev. 2004;30:269–280. doi: 10.1016/j.ctrv.2003.11.001. [DOI] [PubMed] [Google Scholar]
  • 11.Bair E, Tibshirani R. Semi-supervised methods to predict patient survival from gene expression data. PLoS Biol. 2004;2:E108. doi: 10.1371/journal.pbio.0020108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Simon R, Radmacher MD, Dobbin K, et al. Pitfalls in the use of DNA microarray data for diagnostic and prognostic classification. J Natl Cancer Inst. 2003;95:14–18. doi: 10.1093/jnci/95.1.14. [DOI] [PubMed] [Google Scholar]
  • 13.Ramaswamy S, Ross KN, Lander ES, et al. A molecular signature of metastasis in primary solid tumors. Nat Genet. 2003;33:49–54. doi: 10.1038/ng1060. [DOI] [PubMed] [Google Scholar]
  • 14.Weigelt B, Hu Z, He X, et al. Molecular portraits and 70-gene prognosis signature are preserved throughout the metastatic process of breast cancer. Cancer Res. 2005;65:9155–9158. doi: 10.1158/0008-5472.CAN-05-2553. [DOI] [PubMed] [Google Scholar]
  • 15.Simon R. Roadmap for developing and validating therapeutically relevant genomic classifiers. J Clin Oncol. 2005;23:7332–7341. doi: 10.1200/JCO.2005.02.8712. [DOI] [PubMed] [Google Scholar]
  • 16.Li H, Gui J. Partial Cox regression analysis for high-dimensional microarray gene expression data. Bioinformatics. 2004;20(suppl 1):i208–i215. doi: 10.1093/bioinformatics/bth900. [DOI] [PubMed] [Google Scholar]
  • 17.van Houwelingen HC, Bruinsma T, Hart AA, et al. Cross-validated Cox regression on microarray gene expression data. Stat Med. 2006;25:3201–3216. doi: 10.1002/sim.2353. [DOI] [PubMed] [Google Scholar]
  • 18.Pawitan Y, Bjohle J, Wedren S, et al. Gene expression profiling for prognosis using Cox regression. Stat Med. 2004;23:1767–1780. doi: 10.1002/sim.1769. [DOI] [PubMed] [Google Scholar]
  • 19.Davicioni E, Anderson MJ, Finckenstein FG, et al. Molecular classification of rhabdomyosarcoma: Genotypic and phenotypic determinants of diagnosis—A report from the Children's Oncology Group. Am J Pathol. 2009;174:550–564. doi: 10.2353/ajpath.2009.080631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Davicioni E, Finckenstein FG, Shahbazian V, et al. Identification of a PAX-FKHR gene expression signature that defines molecular classes and determines the prognosis of alveolar rhabdomyosarcomas. Cancer Res. 2006;66:6936–6946. doi: 10.1158/0008-5472.CAN-05-4578. [DOI] [PubMed] [Google Scholar]
  • 21.Wachtel M, Dettling M, Koscielniak E, et al. Gene expression signatures identify rhabdomyosarcoma subtypes and detect a novel t(2;2)(q35;p23) translocation fusing PAX3 to NCOA1. Cancer Res. 2004;64:5539–5545. doi: 10.1158/0008-5472.CAN-04-0844. [DOI] [PubMed] [Google Scholar]
  • 22.Wachtel M, Runge T, Leuschner I, et al. Subtype and prognostic classification of rhabdomyosarcoma by immunohistochemistry. J Clin Oncol. 2006;24:816–822. doi: 10.1200/JCO.2005.03.4934. [DOI] [PubMed] [Google Scholar]
  • 23.Allory Y, Matsuoka Y, Bazille C, et al. The L1 cell adhesion molecule is induced in renal cancer cells and correlates with metastasis in clear cell carcinomas. Clin Cancer Res. 2005;11:1190–1197. [PubMed] [Google Scholar]
  • 24.Gavert N, Conacci-Sorrell M, Gast D, et al. L1, a novel target of beta-catenin signaling, transforms cells and is expressed at the invasive front of colon cancers. J Cell Biol. 2005;168:633–642. doi: 10.1083/jcb.200408051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Thies A, Schachner M, Moll I, et al. Overexpression of the cell adhesion molecule L1 is associated with metastasis in cutaneous malignant melanoma. Eur J Cancer. 2002;38:1708–1716. doi: 10.1016/s0959-8049(02)00105-3. [DOI] [PubMed] [Google Scholar]
  • 26.Nagle JA, Ma Z, Byrne MA, et al. Involvement of insulin receptor substrate 2 in mammary tumor metastasis. Mol Cell Biol. 2004;24:9726–9735. doi: 10.1128/MCB.24.22.9726-9735.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pursglove SE, Mackay JP. CSL: A notch above the rest. Int J Biochem Cell Biol. 2005;37:2472–2477. doi: 10.1016/j.biocel.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • 28.Wechsler-Reya RJ, Elliott KJ, Prendergast GC. A role for the putative tumor suppressor Bin1 in muscle cell differentiation. Mol Cell Biol. 1998;18:566–575. doi: 10.1128/mcb.18.1.566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sakamuro D, Prendergast GC. New Myc-interacting proteins: A second Myc network emerges. Oncogene. 1999;18:2942–2954. doi: 10.1038/sj.onc.1202725. [DOI] [PubMed] [Google Scholar]
  • 30.Fan C, Oh DS, Wessels L, et al. Concordance among gene-expression-based predictors for breast cancer. N Engl J Med. 2006;355:560–569. doi: 10.1056/NEJMoa052933. [DOI] [PubMed] [Google Scholar]
  • 31.Nevins JR, Potti A. Mining gene expression profiles: Expression signatures as cancer phenotypes. Nat Rev Genet. 2007;8:601–609. doi: 10.1038/nrg2137. [DOI] [PubMed] [Google Scholar]
  • 32.Xia SJ, Barr FG. Analysis of the transforming and growth suppressive activities of the PAX3-FKHR oncoprotein. Oncogene. 2004;23:6864–6871. doi: 10.1038/sj.onc.1207850. [DOI] [PubMed] [Google Scholar]
  • 33.Sorensen PH, Lynch JC, Qualman SJ, et al. PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: A report from the Children's Oncology Group. J Clin Oncol. 2002;20:2672–2679. doi: 10.1200/JCO.2002.03.137. [DOI] [PubMed] [Google Scholar]
  • 34.Bennicelli JL, Advani S, Schafer BW, et al. PAX3 and PAX7 exhibit conserved cis-acting transcription repression domains and utilize a common gain of function mechanism in alveolar rhabdomyosarcoma. Oncogene. 1999;18:4348–4356. doi: 10.1038/sj.onc.1202812. [DOI] [PubMed] [Google Scholar]
  • 35.Du S, Lawrence EJ, Strzelecki D, et al. Co-expression of alternatively spliced forms of PAX3, PAX7, PAX3-FKHR and PAX7-FKHR with distinct DNA binding and transactivation properties in rhabdomyosarcoma. Int J Cancer. 2005;115:85–92. doi: 10.1002/ijc.20844. [DOI] [PubMed] [Google Scholar]
  • 36.Kelly KM, Womer RB, Sorensen PH, et al. Common and variant gene fusions predict distinct clinical phenotypes in rhabdomyosarcoma. J Clin Oncol. 1997;15:1831–1836. doi: 10.1200/JCO.1997.15.5.1831. [DOI] [PubMed] [Google Scholar]
  • 37.Pappo AS, Anderson JR, Crist WM, et al. Survival after relapse in children and adolescents with rhabdomyosarcoma: A report from the Intergroup Rhabdomyosarcoma Study Group. J Clin Oncol. 1999;17:3487–3493. doi: 10.1200/JCO.1999.17.11.3487. [DOI] [PubMed] [Google Scholar]
  • 38.Qualman SJ, Bowen J, Parham DM, et al. Protocol for the examination of specimens from patients (children and young adults) with rhabdomyosarcoma. Arch Pathol Lab Med. 2003;127:1290–1297. doi: 10.5858/2003-127-1290-PFTEOS. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Supplement
Publisher's Note

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES