Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 AuthorPublished manuscripts OnlineFirst have been peer on reviewed July 7, and 2010 accepted as 10.1158/1078-0432.CCR-10-0284 for publication but have not yet been edited.

Genomic alterations and allelic imbalances are strong prognostic predictors in osteosarcoma

Jan Smida1,2§, Daniel Baumhoer1,3,4§, Michael Rosemann5,6, Axel Walch5, Stefan Bielack7, Christopher Poremba8, Klaus Remberger9, Eberhard Korsching10, Wolfram Scheurlen11, Christian Dierkes12, Stefan Burdach2, Gernot Jundt3,4, Michael J. Atkinson5,6, and Michaela Nathrath1,2

1Clinical Cooperation Group Osteosarcoma, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Germany, 2Department of Pediatrics, Technische Universitaet Muenchen and Pediatric Oncology Center, Germany, 3Institute of Pathology and 4Bone Tumor Reference Center at the Institute of Pathology, University Hospital Basel, Switzerland, 5Institute of Pathology and 6Institute of Radiation Biology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Germany, 7Klinikum Stuttgart Olgahospital, Pediatrics 5 - Oncology, Hematology, Immunology, Germany, 8Institute of Pathology, Heinrich-Heine-University, Duesseldorf, Germany and Center of Histopathology, Cytology and Molecular Diagnostics (CHCMD), Research Park Trier, Germany, 9Institute of Pathology, University of the Saarland, Homburg-Saar, Germany, 10Institute of Bioinformatics, University of Muenster, Germany, 11Cnopfsche Kinderklinik Nuremberg Children’s Hospital, Germany, and 12Institute of Pathology, Justus-Liebig-University, Giessen, Germany § The authors contributed equally to this work

Corresponding author / adress for reprint requests: Michaela Nathrath, Clinical Cooperation Group Osteosarcoma, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany, Phone: +49-89-3187-3797, Fax: +49-89-3187-3381, EMail: [email protected]

Grant support: Jan Smida, Eberhard Korsching and Michaela Nathrath are members of the Translational Sarcoma Research Network supported by the BMBF. Michaela Nathrath is supported by the Wilhelm Sander-Stiftung and the Elterninitiative krebskranke Kinder München e.V. Daniel Baumhoer was supported by the Novartis Foundation, formerly Ciba-Geigy-Jubilee-Foundation.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Running title: Prognostic impact of genomic alterations in osteosarcoma

Key words: osteosarcoma, prognosis, SNP array, CDK4, MYC

Disclosure / Conflict of Interest: We declare that we have no conflict of interest.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Statement of Translational Relevance

Osteosarcoma is a highly aggressive neoplasm with 30-40% of patients dying due to progressive disease, despite of intense multidisciplinary treatment protocols. Up to now, the clinical course of patients can not be anticipated reliably at the time of initial diagnosis and generally the histologically assessed response to neoadjuvant chemotherapy after definitive surgery has to be awaited. Since up to 50% of osteosarcoma patients only poorly respond to neoadjuvant treatment, molecular predictors might help to stratify patients more accurately in distinct prognostic and therapeutic subgroups. In the present study, we propose that genomic alterations, determined by using a chromosomal alteration staging (CAS) system, are strong prognostic predictors in osteosarcoma that can be veryfied at initial biopsy and are superior to the histologic regression grading.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

ABSTRACT

Purpose: Osteosarcoma, the most common primary malignant tumor of bone, is characterized by complex karyotypes with numerous structural and numerical alterations. Despite attempts to establish molecular prognostic markers at the time of diagnosis, the most accepted predictive factor remains the histological evaluation of necrosis after neoadjuvant chemotherapy. The present approach was carried out to search for genome-wide recurrent loss of heterozygosity (LOH) and copy number variations (CNV) that could have prognostic and therapeutic impact for osteosarcoma patients. Experimental Design: Pretherapeutic biopsy samples of 45 osteosarcoma patients were analyzed using Affymetrix 10K2 high-density SNP arrays. Numerical aberrations and allelic imbalances were correlated with the histologically assessed response to therapy and clinical follow-up. Results: The most frequent genomic alterations included amplifications of 6p21 (15.6%), 8q24 (15.6%, harboring MYC) and 12q14 (11.1%, harboring CDK4) as well as LOH of 10q21.1 (44.4%). All these aberrations and the total degree of heterozygosity of each tumor were significantly associated with an adverse outcome of patients and used to define a chromosomal alteration staging (CAS) system with a superior predictive potential compared to the histologic regression grading. Conclusions: Structural chromosomal alterations detected by SNP analysis provide a simple but robust parameter to anticipate response to chemotherapy. The proposed CAS system might therefore help to better predict the clinical course of osteosarcoma patients at the time of initial diagnosis and to adept neoadjuvant treatment in patients resistant to the current protocols.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

INTRODUCTION

Due to advances in therapy over the last decades, long-term survival in osteosarcoma patients with localized disease has improved and now reaches about 65% with multimodal therapy. Nevertheless, 30-40% of the patients die because of tumor progression or relapse (1). Individualized intervention schemes based on the biological characteristics of each tumor have been shown to be successful in other pediatric cancer types e.g. in acute lymphatic leukaemia and in neuroblastoma. Also concerning osteosarcomas, several studies have been carried out to combine common histological parameters with genetic and molecular findings to predict the clinical outcome of patients (2-4). However, the histological response to neoadjuvant chemotherapy (“regression grade”) assessed after definitive surgery is still the gold standard concerning prognostic prediction (5, 6). Osteosarcomas are characterized by highly complex karyotypes and a high frequency of chromosomal copy number changes (3, 7-12). It has already been shown in other tumors, such as gastrointestinal tract carcinomas, that a high complexity of chromosomal instability is correlated with an unfavorable outcome (13). In this study, we describe the use of Affymetrix single nucleotide polymorphism (SNP) arrays in a genome-wide high-resolution approach. Both, loss of heterozygosity (LOH) and variations in DNA copy numbers (CNV), were assayed in order to identify possible genomic fingerprints for the prediction of response to chemotherapy and prognosis. Furthermore, the osteosarcoma genomes were investigated for chromosomal gains and losses to identify candidate as potential therapeutic targets.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

MATERIAL AND METHODS

Tissue samples and patients characteristics Our series of pretherapeutic fresh frozen biopsy samples included 79 osteosarcomas which were numbered consecutively (OS1-OS79). Due to insufficient size or inadequate amount of vital tumor only 45 samples were selected for the present study including osteosarcoma of the extremities (n = 43) and the pelvis (n = 2). In nine cases, tumor and blood samples from the same patient were available. There were 25 males and 20 females with a mean age of 16.5 years (median 14 years; range 4–51 years); 10 patients had lung metastases at the time of initial diagnosis. All patients were treated between 1993 and 2007. Preoperative and postoperative chemotherapy was given according to the protocols of the Cooperative German-Austria-Swiss Osteosarcoma Study Group (reviewed and approved by the appropriate ethics committee) after informed consent. Response to chemotherapy was assessed according to the Salzer- Kuntschik (S-K) histological six-graded scale. SK-grades 1 to 3 (≤ 10% viable tumor cells) were classified as good responders and SK-grades 4 to 6 (> 10% viable tumor cells) as poor responders. Information on histological response was available for 44 patients. Follow-up data (0.6-12.7 years, mean 3.84 years) were available for all patients (Table 1). Additionally, blood samples from 5 healthy donors were used as hybridization quality controls.

Affymetrix 10K2 high-density SNP arrays Genomic DNA was extracted using the QIAamp-Mini DNA extraction kit (Quiagen, Hilden, Germany) from frozen tissue and blood samples. DNA samples were processed according to the standard GeneChip Mapping 10K (V2.0) Xba Assay protocol (Affymetrix Inc., Santa Clara, CA) as suggested by the manufacturer [http://www.affymetrix.com/products/arrays/index.affx]. In brief, 250 ng of DNA was digested with XbaI and ligated to the XbaI adaptor prior to PCR amplification using AmpliTaq Gold (Applied Biosystems, Foster City, CA) and primers that recognize the adapter sequence. PCR-amplified DNA was fragmented using DNAse I, end-labeled with a fluorescent tag and hybridized to the array. Hybridized arrays were processed with an Affymetrix Fluidics Station 450 and fluorescence signals were detected using the Affymetrix GeneChip Scanner 3000.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

The primary hybridization data were processed using Affymetrix GCOS and GTYPE software, yielding the genotype and hybridization intensity for each individual SNP marker. In a second step the Affymetrix® GeneChip® Chromosome Copy Number Analysis Tool (CNAT and GTYPE Version 4.1) was applied to visualize the data corresponding to their chromosomal location and to convert the hybridization intensities into DNA copy numbers (CN). Copy number variations (CNV) with a score below 0.75 were classified as losses and with a score >3 as gains. CNAT also detects genomic regions with unusual or long stretches of contiguous homozygote SNPs and assigns them a score for the likelihood of being caused by a tumor specific loss of heterozygosity (LOH). To distinguish tumor-specific somatic LOH from potential germline homozygosity, a cut-off LOH value of 5 was used. The value varies between 0.01 (no LOH = heterozygous SNP) and 75 (no heterozygous SNP on the entire chromosome arm) and has been explicitly discussed in Affymetrix White Papers. We validated this threshold by comparing regions that showed allelic losses in a small subset of tumor and blood samples. The cut-off LOH value of 5 had the best fit with the LOH data of matched samples and is equivalent to stretches of homozygosity greater than 4 Mbp. A homozygous interval of this length was not observed either in the investigated blood samples or in the genome of more than 100 healthy donors (control- data provided by Affymetrix). All physical positions were determined using the NetAffxTM Analysis Center's UCSC Genomic Query Tool provided by the manufacturer and correspond to the NCBI sequencing database (version GRCh37, hg19). All statistical analyses were done with STATISTICA 4.5 (StatSoft, Tulsa, OK, USA). The correlation analysis performed in this study used Spearman's rank order correlation coefficient. Differences between good and poor responders to chemotherapy were considered significant at P < 0.05 (α-2 sides) using the Wilcoxon-Mann-Whitney rank test.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

RESULTS

We collected frozen pretherapeutic biopsy specimens of osteosarcomas from 45 patients, all but one with a record of the subsequent response to chemotherapy. We used Affymetrix 10K SNP microarrays to assay DNA copy-number changes and LOH. A mean call rate of 88.5% (range 82.71% to 96.2%) resulted in a minimum of 8.200 SNP genotypes per case.

Heterozygosity and LOH In all osteosarcoma samples we observed multiple regions of contiguous SNP homozygosity, ranging in length from 4.7 Mb to 156.6 Mb. These regions were classified by Affymetrix analysis software as areas with LOH (LOH-likelihood score 5 to 70, determined by Affymetrix CNT tool). An appropriate method to estimate the whole-genome LOH frequency is to compare the frequency of SNP genoytpes in a tumor with the population average. All SNPs used in the array are known to be heterozygous in at least 38% of the reference population (data by Affymetrix). The effective heterozygosity in the investigated blood samples as well as in control DNA provided by Affymetrix was on average 34.4%. The percentage of heterozygote markers in the osteosarcoma samples, however, droped to an average of 27% in good responders and even to 24% in poor responders (Fig. 1A). Applying the LOH scoring algorithm as described above this converts into approximately 100, 1200 and 2000 out of 9769 SNPs being homozygote due to a putative allelic loss in blood samples, good-responding tumors and poor-responding tumors, respectively (Fig. 1B). Any LOH in a tumor sample is evident from a reduction of the average heterozygosity over contiguous stretches of a chromosome. LOH accompanied by a copy number loss along an entire chromosome, were found in 22 out of 45 tumors. In all investigated osteosarcomas LOH affected at least 6 with a non-randomly distribution throughout the genome. Preferentially affected were chromosomes 2, 3, 5, 6 10 and chromosome 13, which together harbored 51% of all SNPs scored as LOH.

LOH score and LOH profile as putative prediction factors Response to chemotherapy and metastatic disease are well known prognostic factors that were also shown in our cohort of patients: the response to chemotherapy

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

significantly reflected a favorable outcome (event-free survival in 73% of the good responders compared with 38% of the poor responders, P = 0.005, data not shown) while the presence of metastases represented the most decisively negative prognostic factor (Fig. 1C-D). However, the frequency of LOH was also strongly associated with the patients’ outcome (Fig. 1E). The most striking finding was the unequal distribution of LOH with respect to the histologically evaluated response to chemotherapy: patients with a high LOH-score (more SNPs scored as LOH than average of 1500) significantly more often had a poor response to chemotherapy than patients with a low LOH-score (Wilcoxon-Mann-Whitney rank test, data not shown). The prognostic value of heterozygosity depended on the chromosomal region of the affected SNP. The percental distribution of the LOH values along the genome of all investigated osteosarcomas, stratified by their response to chemotherapy, is shown for each autosomal chromosome in Figure 2A.

LOH regions in relation to response to therapy and relapse The most common LOH locus (in 22/45 cases; 48.9%) was found on a 4.2 Mb region of chromosome 13 (13q14.13-13q14.2; minimal deleted region in positions 45.553.450- 49.731.408; 24 SNPs) harboring among other genes the osteosarcoma associated tumor suppressor RB1. LOH on 13q did not show any correlation to the response to chemotherapy or event-free survival (Fig. 2B and 2E). The region 11p15.1-11p15.4 on chromosome 11 exhibited LOH in 5 separate SNPs- stretches (in 12/44 cases; 27%), the most prominent measuring 3.6 Mb (positions 8.460.319- 11.973.737; 16 SNPs) and harboring among others the genes WEE1, ST5 and LMO1. LOHs of 11p were found significantly (P < 0.001) more often in poor responders than in good responders. However, there was no significant correlation with event-free survival (Fig. 2C and 2F). The region 10q21.1 on (2.5 Mb; positions 55.666.767–58.131.081; 17 SNPs), harboring among others PCDH15 and ZWINT1, showed a very high LOH frequency (21/45 cases; 46.7%) and was significantly more often found in patients with poor response to chemotherapy (P < 0.005). LOH in this region was significantly associated with event-free survival, independent of the absence (P = 0.0108) or presence (0.0089) of primary lung metastases (Fig. 2D and 2G).

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Clusters of Copy Number Variations (CNV) LOH can be associated with DNA-Copy number variations (CNV). Signal intensity data for the quantification of allelic copy numbers changes indicated that 41% of the LOHs found in osteosarcoma genomes were due to allelic losses and gains, including events of duplication or chromosomal loss. The remaining 59% of SNPs showed LOH without CNV. Long stretches of homozygosity were frequently associated with CNVs. To predefine groups, the osteosarcomas were clustered according to their CNV-similarity using the processed CNV score-profiles of all SNPs. It was found that there were three different groups of CNV-similarity on individual SNPs (Fig. 3A; squares A, B and C). The three defined clusters illustrate the expected heterogeneity of osteosarcomas. Medial cluster B conjoins osteosarcomas (22 out of 45) of heterogenic and variable characteristics (short branches indicate low distinguishing potential) and with no obvious relation to response and prognosis; Kaplan-Meier analysis of event-free survival reveals no significant correlation with the clustering. However, the clusters A and C, which represent the both extreme characters of the dendrogram (longest branches indicate the highest dissimilarity with other subclusters), include those osteosarcomas that exhibit in a great majority either uniformity in response to chemotherapy (Fig. 3A, cluster C: poor response) or in frequency of LOH per SNP (Fig. 3B, cluster A: on average 10% of SNPs affected).

Regions of high amplifications CNV analysis enabled a construction of CGH-like profiles (including aneuploidy and common variations on chromosome arms) and the detection of amplified “hot spot” regions. In our cohort, 27 out of 45 osteosarcomas exhibited gains of more than two-fold which affected at least one chromosomal region (more than 5 contiguous SNPs), but only 13 of these osteosarcomas showed high level amplifications (> 3-fold copy number gains). Amplifications were found to be most frequent (10 of 45 osteosarcomas) on 6p12-p21 (Fig. 4A), where three separate hot spot regions could be identified. In one case, an additional amplified region of 0.4 Mb was identified at 6p22 harboring the E2F3 gene. Six SNPs in a row displayed a signal that was on average 16 times higher than the surrounding signals (Fig. 4A).

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Amplification in one or more of the three 6p-regions was found in 10/45 osteosarcomas (22%); the most frequently amplified region (in 6/45 osteosarcomas = 14%) was approximately 4,7 Mb in size, localized between physical positions 40,910.958 and 45,607.978, and included the potential candidate genes CCND3, PTK7 and RUNX2. Amplifications on 6p12-p21 were found in both response groups, namely in 4/23 patients with good and 6/22 patients with poor response. However, event-free survival was significantly (P = 0.018) less common in patients, whose osteosarcomas showed high amplifications in this region (Fig. 4B). The MYC-containing region 8q24.21, which is 0,8 Mbp in size, was found to be amplified in 7 cases, including 5 highly amplified cases. Amplifications were significantly associated with an adverse overall survival (P = 0.01) and event-free survival (P = 0.0003). Three out of 7 patients exhibiting an 8q-amplification had primary pulmonary metastases (ppm). However, even after excluding these 3 patients with ppm from the collective the remaining 4 had an invariable significant association with event-free survival (P = 0.004) and overall survival (P = 0.01) (Fig. 4C). The CDK4-harboring region 12q14, which is 4 Mbp in size, was found to be amplified in 5 cases, including 3 highly amplified cases. All 5 amplifications were associated with primary lung metastases (Fig. 4D) and with relapse (P = 0.07). Additionally, 10 cases had LOH stretches at the same region that were significantly associated with poor event-free survival in the whole collective (P = 0.002) and also in patients without primary metastases (P = 0.011).

Chromosomal alteration staging as predictive factor The association between chromosomal alterations and clinical outcome of patients was investigated using Fisher’s exact test. Therefore three distinct constellations of genomic events were defined and subsequently compared with the Salzer-Kuntschik regression grading system concerning their prognostic impact: CAS1 included tumors with either an above average LOH score (more SNPs scored as LOH than average of 1500) or amplification of 6p, 8p or 12q, CAS2 comprised osteosarcomas with either LOH on 10q or amplification of 6p, 8p or 12q and CAS3 comprehended cases with a combination of 2 out of the following 4 events: LOH on 10q, amplification of 6p, 8q or 12q. Table 2 compares the Salzer-Kuntschik regression grading and the proposed CAS

constellations regarding the number of correctly predicted cases (nC). The occurrence of

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

relapse was used to differentiate between cases with good and poor prognosis. No difference was found between the group of good responders (S-K I-III) and CAS3 (18 correctly predicted cases with no relapse in both groups), but in all other subgroups CAS1-3 were superior in predicting the correct clinical course of patients compared with the S-K score. Since the cut-off of 10% viable tumor cells defines good and poor responders we firstly focussed on all cases showing a S-K grade III or IV, comprising exactly the cases that were close to that cut-off score. Among these cases, CAS3 predicted the clinical course accurately in 80.8% of patients compared with only 65.4% using the S-K regression grading. Also in the group of S-K poor responders (IV-VI), CAS3 predicted more than 25% more patients correctly than the S-K score (i.e. S-K IV-

VI nC = 61.9% vs nC = 90.5% in CAS3). In Figure 4E-G the proposed CAS categories were evaluated concerning event-free survival and compared with the Salzer-Kuntschik regression grading (Kaplan-Meier analysis). CAS3 unequivocally displays a better prediction of the clinical outcome of patients compared with the histologically assessed response to therapy. Since metastases at the time of initial diagnosis are the most decisive prognostic factor in osteosarcoma patients (Fig. 1C-D) we omitted these cases (n = 10) in a separate calculation. In Figure 4H-K the proposed CAS categories were again compared with the Salzer-Kuntschik score and still show a highly significant predictive impact. Nevertheless, there were two cases with primary metastases that were CAS1-3 negative and clinically showed a complete remission following therapy. One case is free of disease since 4.77 years, the other case only since 0.76 years. Although nine month of follow-up is definitely not long enough to consider a patient cured, there is at least one patient with metastatic disease at the time of diagnosis that was correctly classified as CAS negative. The remaining eight cases with primary metastases were all CAS positive and relapsed (n = 1) or died of disease (n = 7) subsequently. Chromosomal alteration staging therefore seems to be meaningful also in patients with primary metastases. We did not find statistically significant correlations between osteosarcoma subtypes and genomic alterations, but at least one interesting trend: 11/45 osteosarcomas demonstrated at least partial chondroblastic differentiation and 8/11 were classified as poor responders according to the Salzer-Kuntschik grade. However, 4/8 poor responders are currently free of disease (range of follow-up 1,56-5,47 years, mean 3.20

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

years) and, thus, were prognostically misclassified. Interestingly, all these cases were CAS negative contrary to the remaining 4/8 poor responders that indeed revealed a poor clinical outcome and all turned out to be CAS positive.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

DISCUSSION

Although the understanding of the molecular pathogenesis of osteosarcoma has advanced in the last two decades, risk assessment continues to be based mainly on clinical and histopathological parameters (14-16). Many studies have been conducted in recent years in order to establish molecular markers that might help to identify patients for whom maximal therapy is necessary and others for whom therapy of reduced intensity is sufficient to achieve long-term survival. So far, such molecular markers have not been identified. The present study was undertaken to search for genes or chromosomal locations with potential pathogenetic, prognostic and therapeutic impact in a genome-wide approach.

LOH patterns in osteosarcoma In the vast majority of the genomes of the investigated osteosarcomas we observed LOHs and CNVs indicating a high degree of chromosomal aberrations and allelic imbalances. Significantly more chromosomal variations, especially LOHs, were observed in tumors with a poor response compared to tumors with a good response to preoperative chemotherapy. Furthermore, patients with tumors showing above average LOH-scores developed recurrent disease more often than patients with tumors showing low LOH-scores. A correlation between chromosomal imbalances and the prognosis of patients has recently been shown not only in osteosarcomas, but also in other tumors, including gastric carcinomas, however the biological basis of these observations is poorly understood (13, 17, 18). The high LOH scores in osteosarcomas with a poor response to chemotherapy found in our study might represent an imbalanced loss of various genes involved in cell cycle and apoptosis regulation and therefore might reflect a more aggressive phenotype with a potential survival advantage against chemotherapy. The locus most frequently affected by LOH in our study was found on a 4.2 Mb region of chromosome 13, harboring the tumor suppressor gene RB1. The RB1-LOH rate of 43% in our study was comparable to the rate of 37.2% to 39% published in the literature (19, 20). Allelic imbalances of the RB1 locus have previously been reported in osteosarcoma but their predictive impact remains controversial. Earlier studies revealed high LOH rates for osteosarcoma at the 13q14 locus and the authors found them to be associated

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

with a poor prognosis (20, 21). More recently, Heinsohn et al. were not able to confirm this prognostic impact, which is in accordance with our findings (19). Remarkably, the LOH region on chromosome 10q21.1, harboring PCDH15 and ZWINT1, was more important in respect of prognostic discrimination (Fig. 2D and 2G). The observed LOH frequency was equivalent to those of the RB1 locus, but occurrence of LOH in this region was adversely correlated with event-free and overall survival. Another region with a recurring LOH pattern in 22% of the investigated cases was found on chromosome 12q13-14 harboring CDK4, which was described in other studies before (16, 22). In our study, we remarkably did not only find amplifications of this region but also high score LOH-stretches which were significantly correlated with relapse. Only recently, the deletion of the 9p region containing the CDKN2A gene has been reported to represent an early event in mouse models for osteosarcoma development and to constitute an independent factor for adverse clinical outcome of osteosarcoma patients (23, 24). Due to limited resolution of the 10K2 high-density SNP arrays used, our data did not allow to determine deletions of the CDKN2A gene but detected LOHs of the respective locus in 5/45 cases (11%), which is in agreement with the literature. However, we found no correlation to clinico-pathological parameters including the clinical outcome of patients. Freeman and colleagues reported the common event of copy number gains in the EGFR and copy number losses in the PTEN gene and therefore suggested a dysregulated PI3K-AKT/mTOR pathway to play a role in the development of osteosarcoma (25). In our study we identified LOHs in the region of the EGFR gene in 13/45 cases (29%) and of the PTEN gene in 6/45 cases (13%). In agreement with the study of Freeman et al. we did not find correlations to clinico- pathological parameters. Also the region 3q13.31, harboring the LSAMP gene, has recently been reported to be commonly affected by chromosomal alterations in osteosarcoma. These findings, predominantly representing deletions, were correlated with disease progression and poor survival (26, 27). Due to sufficient resolution of the respective region, we were able to detect deletions in 15/45 cases (33%) in our series, 5 of which additionally showed LOHs in the flanking DNA. Additionally, we found four cases with LOH that did not show coincidental deletion of 3q13.31. However, although not statistically significant, the deletion of 3q13.31 showed a trend towards adverse outcome in our series, which is in agreement with the literature (26, 27).

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

CNV and Aneuploidy To estimate the differentiation power of CNV values determined in our study we constructed clusters of similarity (Fig. 3). The resulting tree revealed three major similarity groups and one outlier (OS49). Two groups (denoted A and C) covered osteosarcomas that exhibited a good correlation between LOH and the response to chemotherapy. The remaining cases (group B, harboring the majority of patients with primary metastases) represented another subgroup of osteosarcomas which lacked this concordance. Thus, a definite classification of osteosarcomas is not feasible on the basis of the CNV pattern, most probably due to the high heterogeneity of these tumors. Suggested proclivity for genomic instability in cancer cells is reflected by the degree of aneuploidy (28). It has been suggested that imbalances affecting thousands of genes as caused by long-range chromosomal gains or losses may be an independent contributor to carcinogenesis (29, 30). Frequent amplification of centromeres or abnormalities in spindle apparatus development leading to missegregation of chromosomes has been observed in numerous types of malignant tumors and is considered as the major contributing factors for chromosome instability in cancer cells (31). Interpretation and combination of our LOH- and CNV-data allow an overview of frequencies of which chromosomes or chromosomal parts are affected by allelic duplications or losses (virtual karyotyping). Aneuploidy is very common in osteosarcomas, only 2 samples within our collective showed a normal karyotype without aneuploidy. On average 5.5 chromosomes were affected in every sample (median 6), with a maximum of 16 chromosomes in the karyotype of one patient. Aneuploidy is a common characteristic of tumors and has been proposed to drive tumor progression (32-34). Our results show that high level aneuploidy in osteosarcomas is related to poor response to chemotherapy, probably reflecting the aggressiveness of these tumors.

Amplification hotspots Chromosome 6p is often affected by copy number gains in several types of cancer, as revealed by numerous CGH studies (35, 36). The most commonly amplified genomic interval of 6p21–p23 harbors among others several candidate genes: angiogenesis- associated VEGFA, cell cycle-associated E2F3, CCND3, RUNX2 and carcinoma- associated PTK7. These genes are directly or indirectly involved in the pathogenesis and pathways of malignant tumors and amplifications have also been described in a

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

high percentage of osteosarcomas (4, 11, 12, 17, 37-39). In the present study, we found high amplification spots in 24% of the investigated tumors (Fig. 4B) that were significantly correlated with a poor event-free survival. Our data imply a complex pattern of imbalances; there is not a single amplicon covering the whole central region of 6p, instead we found a few smaller hot spot regions harboring CCND3, RUNX2 and VEGFA. These genes have already been found to be overexpressed in osteosarcomas (38) and high expression of VEGFA has been correlated with a poor prognosis (40, 41). Interestingly, the cell-cycle-associated E2F3, a downstream target of RB1, which has been found amplified and overexpressed in other tumors e.g. in urothelial cancer (42), was the only gene located within a small amplified spot suggesting an alternative way to interfere with the RB1-pathway. Amplifications found on 8q24.21 (MYC) were significantly correlated with poor event- free survival, independent of the presence of primary metastases. Only one patient of our study harboring this amplification is still event-free (short follow-up of only 1.2 years up to now). Prior investigations on mRNA expression of c-MYC in osteosarcomas showed a correlation between c-myc overexpression and event-free survival (43), supporting the suggested unfavorable prognostic value of the MYC gene overexpression. The authors found c-MYC overexpression in 42% of osteosarcomas, more than two times more frequent than in our group of tumors (15.6%). These findings implicate that DNA copy-number gains are not the only possible mechanisms to regulate c-MYC expression. MYC amplification is already known as a poor prognostic marker in other tumors e.g. neuroblastomas and lead to therapy stratification. The distinct prognostic value of MYC amplification in our study should be verified prospectively. Amplification of 12q14 has already been shown in osteosarcomas (44). This region harbors CDK4, an important co-factor in mechanisms regulating cell-cycle progression, and MDM2, often reported to be co-amplified with CDK4. Amplification of the genomic interval containing MDM2 was not found in our study, a strong hint in favor of two independent amplification events (45, 46). Amplifications on 12q14 were found in 11% of the investigated tumors, but exclusively in patients with primary pulmonary metastases, suggesting a potential role of CDK4 in developing a metastatic phenotype at least in a subset of osteosarcomas.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Chromosomal alteration staging system as predictive factors Our study reports one of the first uses of a SNP array for genome-wide screening in osteosarcoma, a tumor type that is known for its complex genomic alterations. This complexity hampers the identification of genes in all genome-wide approaches, but confers a clear advantage for studies that combine LOH and CNV data concurrently. With the powerful method of SNP array analysis, we identified a robust and simple marker, the overall genomic instability (the overall LOH score), to predict response to chemotherapy at the time of diagnosis. Additionally, we have shown that this tool is able to find relevant genes for prognosis (MYC, CDK4) and to identify new potential target genes (E2F3, ZWINT-1), which will be subject to ongoing studies. Based on the detailed analysis of chromosomal alterations we were able to define a staging system (CAS) that suggests high superiority in predicting the clinical outcome of patients compared with the histologically assessed regression grading. Especially the combination of two different events of chromosomal instability (LOH on 10q, amplification of 6p, 8q or 12q), designated CAS3, was significantly more reliable in differentiating patients with good and poor prognosis, even in the S-K groups near the cut-off of 10% viable tumor cells (S-K groups III and IV, defining good and poor responders). Notably, our proposed CAS system is able to predict the prognosis of osteosarcoma patients at the time of initial diagnosis, whereas the histologic assessment is possible not before the end of neoadjuvant treatment. Since the currently used regimens include high-dose chemotherapy for ten weeks prior to definitive surgery, knowledge of the achievable effect of this treatment at initial diagnosis might lead to refinement and modification of the therapy-schedule. Furthermore, patients who do not benefit from the current chemotherapy protocols could be spared its associated short-term and long-term side effects. Importantly, the proposed CAS system is applicable for both osteosarcoma patients with and without primary metastases as was shown by omitting the patients with metastatic disease at the time of diagnosis (Fig. 4 H-K). Even in the subset of cases with initial metastases the CAS system was able to detect CAS negative cases that are currently free of disease. The practicability of CAS in the routine diagnostic procedure has to be proven. At present, we are developing a simplified test based on PCR-SNP assays, whose analysis is technologically feasible in a modern diagnostic laboratory. It will be important to validate the findings reported here in prospective clinical trials and to evaluate the

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

usefulness of the chromosomal markers identified here for strategic treatment decisions in osteosarcoma.

Acknowledgements We thank Nina Weber for excellent technical assistance.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

LITERATURE

1. Bielack SS, Kempf-Bielack B, Delling G, et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol 2002;20:776-90. 2. Al-Romaih K, Bayani J, Vorobyova J, et al. Chromosomal instability in osteosarcoma and its association with centrosome abnormalities. Cancer Genet Cytogenet 2003;144:91-9. 3. Man TK, Chintagumpala M, Visvanathan J, et al. Expression profiles of osteosarcoma that can predict response to chemotherapy. Cancer Res 2005;65:8142- 50. 4. Squire JA, Pei J, Marrano P, et al. High-resolution mapping of amplifications and deletions in pediatric osteosarcoma by use of CGH analysis of cDNA microarrays. Genes Chromosomes Cancer 2003;38:215-25. 5. Salzer-Kuntschik M, Brand G, Delling G. [Determination of the degree of morphological regression following chemotherapy in malignant bone tumors]. Pathologe 1983;4:135-41. 6. Salzer-Kuntschik M, Delling G, Beron G, Sigmund R. Morphological grades of regression in osteosarcoma after polychemotherapy - study COSS 80. J Cancer Res Clin Oncol 1983;106 Suppl:21-4. 7. Bayani J, Zielenska M, Pandita A, et al. Spectral karyotyping identifies recurrent complex rearrangements of chromosomes 8, 17, and 20 in osteosarcomas. Genes Chromosomes Cancer 2003;36:7-16. 8. Boehm AK, Neff JR, Squire JA, Bayani J, Nelson M, Bridge JA. Cytogenetic findings in 36 osteosarcoma specimens and a review of the literature. Pediatric Pathology and Molecular Medicine 2000;19:359-76. 9. Bridge JA, Nelson M, McComb E, et al. Cytogenetic findings in 73 osteosarcoma specimens and a review of the literature. Cancer Genet Cytogenet 1997;95:74-87. 10. Zielenska M, Bayani J, Pandita A, et al. Comparative genomic hybridization analysis identifies gains of 1p35 approximately p36 and chromosome 19 in osteosarcoma. Cancer Genet Cytogenet 2001;130:14-21.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

11. Man TK, Lu XY, Jaeweon K, et al. Genome-wide array comparative genomic hybridization analysis reveals distinct amplifications in osteosarcoma. BMC Cancer 2004;4:45. 12. Ozaki T, Schaefer KL, Wai D, et al. Genetic imbalances revealed by comparative genomic hybridization in osteosarcomas. Int J Cancer 2002;102:355-65. 13. Doak SH. Aneuploidy in upper gastro-intestinal tract cancers--a potential prognostic marker? Mutat Res 2008;651:93-104. 14. Bacci G, Longhi A, Versari M, Mercuri M, Briccoli A, Picci P. Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy: 15-year experience in 789 patients treated at a single institution. Cancer 2006;106:1154-61. 15. Clark JC, Dass CR, Choong PF. A review of clinical and molecular prognostic factors in osteosarcoma. J Cancer Res Clin Oncol 2008;134:281-97. 16. Sandberg AA, Bridge JA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors: osteosarcoma and related tumors. Cancer Genet Cytogenet 2003;145:1-30. 17. Selvarajah S, Yoshimoto M, Ludkovski O, et al. Genomic signatures of chromosomal instability and osteosarcoma progression detected by high resolution array CGH and interphase FISH. Cytogenet Genome Res 2008;122:5-15. 18. Williams L, Somasekar A, Davies DJ, et al. Aneuploidy involving chromosome 1 may be an early predictive marker of intestinal type gastric cancer. Mutat Res 2009. 19. Heinsohn S, Evermann U, Zur Stadt U, Bielack S, Kabisch H. Determination of the prognostic value of loss of heterozygosity at the retinoblastoma gene in osteosarcoma. Int J Oncol 2007;30:1205-14. 20. Patino-Garcia A, Pineiro ES, Diez MZ, Iturriagagoitia LG, Klussmann FA, Ariznabarreta LS. Genetic and epigenetic alterations of the cell cycle regulators and tumor suppressor genes in pediatric osteosarcomas. J Pediatr Hematol Oncol 2003;25:362-7. 21. Feugeas O, Guriec N, Babin-Boilletot A, et al. Loss of heterozygosity of the RB gene is a poor prognostic factor in patients with osteosarcoma. J Clin Oncol 1996;14:467-72. 22. Tang N, Song WX, Luo J, Haydon RC, He TC. Osteosarcoma development and stem cell differentiation. Clin Orthop Relat Res 2008;466:2114-30.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

23. Mohseny AB, Szuhai K, Romeo S, et al. Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2. J Pathol 2009;219:294-305. 24. Ottaviano L, Schaefer KL, Gajewski M, et al. Molecular characterization of commonly used cell lines for bone tumor research: a trans-European EuroBoNet effort. Genes Chromosomes Cancer 2010;49:40-51. 25. Freeman SS, Allen SW, Ganti R, et al. Copy number gains in EGFR and copy number losses in PTEN are common events in osteosarcoma tumors. Cancer 2008;113:1453-61. 26. Pasic I, Shlien A, Durbin AD, et al. Recurrent focal copy-number changes and loss of heterozygosity implicate two noncoding RNAs and one tumor suppressor gene at chromosome 3q13.31 in osteosarcoma. Cancer Res 2010;70:160-71. 27. Yen CC, Chen WM, Chen TH, et al. Identification of chromosomal aberrations associated with disease progression and a novel 3q13.31 deletion involving LSAMP gene in osteosarcoma. Int J Oncol 2009;35:775-88. 28. Duesberg P, Rausch C, Rasnick D, Hehlmann R. Genetic instability of cancer cells is proportional to their degree of aneuploidy. Proc Natl Acad Sci U S A 1998;95:13692-7. 29. Fukasawa K. Centrosome amplification, chromosome instability and cancer development. Cancer Lett 2005;230:6-19. 30. Shi Q, King RW. Chromosome nondisjunction yields tetraploid rather than aneuploid cells in human cell lines. Nature 2005;437:1038-42. 31. Chi YH, Jeang KT. Aneuploidy and cancer. J Cell Biochem 2007;102:531-8. 32. Greenman C, Stephens P, Smith R, et al. Patterns of somatic mutation in human cancer genomes. Nature 2007;446:153-8. 33. Weaver BA, Cleveland DW. Does aneuploidy cause cancer? Curr Opin Cell Biol 2006;18:658-67. 34. Weaver BA, Cleveland DW. Aneuploidy: instigator and inhibitor of tumorigenesis. Cancer Res 2007;67:10103-5. 35. Forus A, Weghuis DO, Smeets D, Fodstad O, Myklebost O, Geurts van Kessel A. Comparative genomic hybridization analysis of human sarcomas: II. Identification of novel amplicons at 6p and 17p in osteosarcomas. Genes Chromosomes Cancer 1995;14:15-21.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

36. Santos GC, Zielenska M, Prasad M, Squire JA. Chromosome 6p amplification and cancer progression. J Clin Pathol 2007;60:1-7. 37. Kasugai Y, Tagawa H, Kameoka Y, Morishima Y, Nakamura S, Seto M. Identification of CCND3 and BYSL as candidate targets for the 6p21 amplification in diffuse large B-cell lymphoma. Clin Cancer Res 2005;11:8265-72. 38. Lu XY, Lu Y, Zhao YJ, et al. Cell cycle regulator gene CDC5L, a potential target for 6p12-p21 amplicon in osteosarcoma. Mol Cancer Res 2008;6:937-46. 39. Lau CC, Harris CP, Lu XY, et al. Frequent amplification and rearrangement of chromosomal bands 6p12-p21 and 17p11.2 in osteosarcoma. Genes Chromosomes Cancer 2004;39:11-21. 40. Huang Y, Lin Z, Zhuang J, Chen Y, Lin J. Prognostic significance of alpha V integrin and VEGF in osteosarcoma after chemotherapy. Onkologie 2008;31:535-40. 41. Sulzbacher I, Birner P, Trieb K, Lang S, Chott A. Expression of osteopontin and vascular endothelial growth factor in benign and malignant bone tumors. Virchows Arch 2002;441:345-9. 42. Hurst CD, Tomlinson DC, Williams SV, Platt FM, Knowles MA. Inactivation of the Rb pathway and overexpression of both isoforms of E2F3 are obligate events in bladder tumours with 6p22 amplification. Oncogene 2008;27:2716-27. 43. Gamberi G, Benassi MS, Bohling T, et al. C-myc and c-fos in human osteosarcoma: prognostic value of mRNA and expression. Oncology 1998;55:556-63. 44. Benassi MS, Molendini L, Gamberi G, et al. Alteration of pRb/p16/cdk4 regulation in human osteosarcoma. Int J Cancer 1999;84:489-93. 45. Berner JM, Forus A, Elkahloun A, Meltzer PS, Fodstad O, Myklebost O. Separate amplified regions encompassing CDK4 and MDM2 in human sarcomas. Genes Chromosomes Cancer 1996;17:254-9. 46. Lopes MA, Nikitakis NG, Ord RA, Sauk J, Jr. Amplification and protein expression of chromosome 12q13-15 genes in osteosarcomas of the jaws. Oral Oncol 2001;37:566-71.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

FIGURE LEGENDS

Figure 1: Affymetrix 10K2 based DrawBoxPlot-analysis of 44 osteosarcoma patients grouped according to their Salzer-Kuntschik (S-K) score in good and poor responders (24 patients with S-K score I-III vs 21 patients with S-K score IV-VI), A: heterozygosity in % SNPs; B: amount of SNPs scored as LOH (9.769 SNPs analyzed); Kaplan-Meier analyses comparing patients with and without primary pulmonary metastases (ppm) show highly significant differences concerning overall survival (C, p=0.00003) and event-free survival (D, p=0.0002); E: Kaplan-Meier analysis comparing patients with and without high LOH scores demonstrate a trend towards adverse outcome for patients with high LOH scores (patients with ppm were excluded in dotted and included in solid lines).

Figure 2: A: The chromosomal pattern of genome-wide LOH probability derived by SNP arrays in 44 osteosarcomas (proportional sized probe sets for each chromosome). Each chromosome illustration shows the average percentage of LOH affected SNPs, calculated for the good (left greenish column) and the poor (right reddish column) response to chemotherapy patient-group, respectively. In the heatmap-diagram, cold colors represent low, hot colors high frequencies of LOH; B-D: LOH distribution in osteosarcomas on chromosomal regions. Average percentage of LOH affected SNPs was calculated separately for patients with poor (black color) and good (grey color) response to chemotherapy. B: chromosome 13 contained a region on 13q14.2 – harboring the RB1 gene – of high LOH frequency that affected 22/44 osteosarcomas (50%); C: 5 distinct regions (marked by red colored bars) on chromosome 11p15.1-4 (marked by red dots) found in 12/44 osteosarcomas (27%) strongly discriminated between poor and good response to chemotherapy; D: chromosome 10q21.1 contained a region with LOH in 21/44 osteosarcomas (47%) that also decisively distinguished between good and poor responders; E-G: Kaplan-Meier analysis for event-free survival

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(the patients with ppm were excluded in dotted and included in solid lines): no significant correlation was found between LOH on 13q14.2 and relapse (E, p=0.3) or between LOH on 11p15.1-4 and relapse (F, p=0.8); between LOH on 10q21.1 and relapse a highly significant correlation was detected (G), independent of including (p=0.0089) or excluding (p=0.0108) ppm patients.

Figure 3: CNV-clusters display the similarity among the entire copy number profile of the osteosarcomas. Numbers indicate individual osteosarcomas. Grey and black bars indicate good and poor response to chemotherapy, respectively. Bars with a red margin indicate patients with primary lung metastases. Three different similarity branches are discriminable (clusters A-C); the only non-clustering sample is osteosarcoma 49. A shows the distribution of the prognostic characteristics defined by histological response and primary lung metastases displayed at the CNV cluster tree. The similarity cluster C includes a high percentage of poor responders (7/8, 87%), small branches in A and B include exclusively osteosarcomas with good response; B shows the distribution of defined LOH at the same CNV-cluster. Remarkably, many osteosarcomas grouped in cluster A (12/16 = 75%) display a low frequency of LOH affected SNPs (<10%).

Figure 4: A: Chromosomal region 6p12-p21 spans a stretch of nearly 25 Mb between the positions 54741994 (6p12) and 30200689 (6p21). High amplification patterns in osteosarcomas found on chromosome 6 showed three distinct loci of amplification with a mean peak of frequency in a 5 Mb stretch between positions 40910958 and 45607978. The additional single event of high-amplification found on 6p22 (at positions 19921598-20345022) harbors the gene E2F3; B-D: high-amplification regions in osteosarcomas – Kaplan-Meier analysis for event-free survival (the patients with ppm were excluded in dotted and included in solid lines); B: 6p12-p21: amplification was significantly associated with poor event-free survival (p=0.018); C: 8q24.21: all patients affected by

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

amplification suffered from relapse within 2 years after initial diagnosis, irrespective of including (p=0.004) or excluding (p=0.0003) ppm patients; D: 12q14: all five patients affected by amplification showed primary metastases; there was a trend towards event-free survival (p=0.07); E-G: Kaplan-Meier analysis for event-free survival comparing the Salzer- Kuntschik regression grading (dotted lines) and the proposed CAS systems (solid lines): E: CAS1, F: CAS2 and G: CAS3; Kaplan-Meier analysis for event-free survival comparing the Salzer-Kuntschik regression grading (dotted lines) and the proposed CAS systems (solid lines) only for patients without primary metastases: H: CAS1, I: CAS2 and K: CAS3.

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A B 35% 6000

25% 3000

15% 0 Controls good poor Controls good poor

C ppm D ppm E LOH-score 1 1 1

n = 35 0,8 0,8 0,8 Below 1500 LOH n = 35 n = 22 (- 4 ppm) 0,6 0,6 0,6

0,4 0,4 survival S(t) 0,4 Above 1500 LOH

n = 10 free survival S(t) n = 24 (- 6 ppm) Probabilty of event n = 10 Probabilty of overall 0,2 0,2 0,2

0 2 4 6 8 10 12 0 2 4 6 8 10 12 0 2 4 6 8 10 12 years

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. A

1 2 3 4 5 6 7 8 9 101112 %SNP scored as LOH 0%

25%

50% 21 OS 21 poor response 13 14 15 16 17 18 19 20 21 22 responseOS 23 good

Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. C B q14.2 p15.4 p15.1 60% 60%

40% 40%

20% 20%

q31.1 q34 p15.4 p13 p12 RB1 Chromosome 13q Chromosome 11p D q21.1 60%

40%

20%

1 18 35p14 52p13 69 86 103 120 137 154 171 188 205 222q21.1 239 256q21.3 273 290 307 324 341 358 375 392 409 426q25.1 443 460 477 494 511 528

Chromosome 10 PCDH15 ZWINT-1

Average LOH in % of SNPs, good response Average LOH in % of SNPs, poor response

E LOH-score in 13q14.13-14.2 F LOH-score in 11p15.1-15.4 G LOH-score in 10q21.1 1 1 1 EFS EFS 0,8 n = 26(-5 ppm) 0,8 n = 31(-6 ppm) 0,8 EFS 0,6 0,6 0,6 n = 25(-5 ppm)

0,4 0,4 0,4

survival S(t) relapse relapse n = 20 (-5 ppm) n = 15 (-4 ppm) 0,2 0,2 0,2 relapse

Probability of event free n = 21 (-5 ppm) 0 0 2 4 6 8 10 12 0 2 4 6 8 10 12 0 2 4 6 8 10 12 years

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. B A Height Height

Downloaded from 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.4 Author manuscriptshavebeenpeerreviewedandacceptedforpublicationbutnotyetedited. A 41 41 A

53 53 Author ManuscriptPublishedOnlineFirstonJuly7,2010;DOI:10.1158/1078-0432.CCR-10-0284 55 55 21 21 48 48 37 37 clincancerres.aacrjournals.org 38 38 12 12 16 16 30 30 29 29 23 23 24 24 27 27 76 76 77 77 B 01 01 B 66 66 19 19 02 02

on September 30, 2021. © 2010American Association for Cancer 60 60 15 15 Research. 63 63 22 22 61 61 62 62 51 51 08 08 75 75 50 50 46 46 59 59 47 47 73 73 79 79 65 65 14 14 03 03 C 17 17 C 28 28 %LOH/SNP

26 Response 26 25 25 32

10%-20% 32 > 30% good metastasis Primary poor metastasis Primary 0-10% 20%-30% 34 34 31 31 33 33 49 49 A 6p Amplifications NoProbe Physical size genes Set Position 25 - 24 - SNP_A-1516084 19921598 423424 bp E2F3 23 - SNP_A-1509639 20345022 SNP_A-1511850 22502049 2880575 bp HDGFL1, GMNN 22 - SNP_A-1518505 25382624 SNP_A-1510234 30200686

21.3 - SNP_A-1514645 36901313 21.2 - SNP_A-1513942 40910958 4697020 bp CCND3, RUNX2 SNP_A-1518706 45607978 21.1 - SNP_A-1516564 46437363

12 - SNP_A-1510836 53540169 5184633 bp KLHL31, ZNF45 11.2 - SNP_A-1510328 54741994 SNP_A-1517757 58724802 11.1 -

B Amplificationon 6p12-21 C Amplificationon 8q24.21 D Amplificationon 12q14 1 1 1 Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peerEFS reviewed and accepted for publication but have not yet beenEFS edited. EFS 0,8 n = 36(-5 ppm) 0,8 n = 39(-7 ppm) 0,8 n = 41

0,6 0,6 0,6

0,4 0,4

survival S(t) 0,4 relapse relapse relapse n = 10 (-5 ppm) n = 7 (-3 ppm) 0,2 0,2 0,2 n = 5 Probabilityofevent free

0 0 2 4 6 8 10 12 0 2 4 6 8 10 12 0 2 4 6 8 10 12 years

E CAS1 staging F CAS2 staging G CAS3 staging 1 1 1 n = 29 n = 26 n = 17 0,8 0,8 0,8

0,6 0,6 0,6 P=6.3x10-6 0,4 0,4 0,4 survival S(t)

0,2 0,2 0,2 Probabilityofevent free P=0.02 P=0.006 0 0 2 4 6 8 10 12 0 2 4 6 8 10 12 0 2 4 6 8 10 12 years

H CAS1 staging I CAS2 staging K CAS3 staging 1 1 1

0,8 n = 21 0,8 n = 18 0,8 n = 9

0,6 0,6 0,6 P=0.0002 0,4 0,4 0,4 survival S(t)

0,2 0,2 0,2 Probabilityofevent free P=0.06 P=0.04 0 0 2 4 6 8 10 12 0 2 4 6 8 10 12 0 2 4 6 8 10 12 years

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 1: Patient characteristics Characteristic Number of patients (n = 45) Age at diagnosis (years) range 4-51 mean 16.5 median 14 Gender (n) male 25 female 20 Blood samples available (n) number 9 Site of primary tumor (n) femur 27 tibia 8 humerus 6 pelvis 2 fibula 1 unknown 1 Histologic differentiation (n) osteoblastic 28 chondroblastic 11 fibroblastic 4 teleangiectatic 2 Metastasis (n) lung 10 Follow up (years) range 0,6-12,7 mean 3.84

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Clinical outcome (n) CR 27 CR-lost 2 AWD-lost 3 DOD 13 Response to chemotherapy (n)

S-K grade I 1 S-K grade II 10 S-K grade III 12 good 23 S-K grade IV 14 S-K grade V 6 S-K grade VI 1 poor 21 no S-K available 1 Abbreviations: CR = complete remission, lost = lost of follow up, AWD = alive with disease, DOD = dead of disease; S-K = Salzer-Kuntschik, grade I = no residual viable tumor, II = solitary viable tumor cells, III = < 10% viable tumor, IV = 10-50% viable tumor, V = 50-80% viable tumor, VI = > 80% viable tumor

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 2: Correlation between the Salzer-Kuntschik regression grading and the proposed chromosomal alteration staging systems

nC

above average LOH-score CAS1 CAS2 CAS3 All cases n = 45 28 (62,2%) 30 (66,6%) 31 (68,8%) 38 (84,4%) nR = 18 ns p=0,0053 p=0,0049 ns (40%) S-K I-III n = 23 nR = 5 11 (47,8%) 11 (47,8%) 13 (56,5%) 18 (78,3%) (21,7%) ns ns ns ns nC = 18 (78,3%) S-K III-IV n = 26 nR = 9 15 (57,7%) 18 (69,2%) 20 (76,9%) 21 (80,8%) (34,6%) ns p=0,0243 p=0,0045 p=0,0008 nC = 17 (65,4%) S-K IV-VI n = 21 16 (76,2%) 18 (85,7%) 17 (81,0%) 19 (90,5%) nR = nC = 13 p=0,0408 p=0,0028 p=0,0117 p=0,0005 (61,9%)

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

S-K V-VI n = 7 6 (85,7%)* 6 (85,7%)* 6 (85,7%)* 7 (100%)* nR = nC = 5 (71,4%)

Abbreviations: nC = number of cases with correct prediction of the clinical course, nR = number of cases with relapse, ns = not significant, * = insufficient case numbers for statistics

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 7, 2010; DOI: 10.1158/1078-0432.CCR-10-0284 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Genomic Alterations and Allelic Imbalances are Strong Prognostic Predictors in Osteosarcoma

Jan Smida, Daniel Baumhoer, Michael Rosemann, et al.

Clin Cancer Res Published OnlineFirst July 7, 2010.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-10-0284

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2010/07/07/1078-0432.CCR-10-0284. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research.