Purpose: In osteosarcoma, aggressive preoperative and postoperative multidrug chemotherapy given to all patients has improved patient survival rate to the present level of ∼60%. However, no tumor marker is available that reliably can identify those patients who will or will not respond to chemotherapy.

Experimental Design: In an attempt to find leads to such markers, we have obtained microarray gene expression profiles from a panel of 10 different human osteosarcoma xenografts and related the results to their sensitivity to ifosfamide, doxorubicin, and cisplatin.

Results: The expression data identified genes with highly significant differential expression between poor and good responder xenografts to the three different drugs: 85 genes for doxorubicin, 74 genes for cisplatin, and 118 genes for ifosfamide. Technical validation with quantitative reverse transcription-PCR showed good correlation with the microarray expression data. Gene Ontology–guided analysis suggested that properties of the poorly responsive xenografts were resistance to undergo programmed cell death and, particularly for ifosfamide, a drive toward dedifferentiation and increased tumor aggressiveness. Leads toward metabolic alterations and involvement of mitochondrial pathways for apoptosis and stress response were more prominent for doxorubicin and cisplatin. Finally, small interfering RNA–mediated gene silencing of IER3 and S100A2 sensitized the human osteosarcoma cell line OHS to treatment with 4-hydroperoxyifosfamide.

Conclusions: The expression profiles contained several novel biomarker candidates that may help predict the responsiveness of osteosarcoma to doxorubicin, cisplatin, and ifosfamide. The potential of selected candidates will be further validated on clinical specimens from osteosarcoma patients. (Clin Cancer Res 2009;15(23):7161–9)

Translational Relevance

This article reports gene expression profiles that distinguish human osteosarcoma xenografts that respond poorly from those showing good response to doxorubicin, cisplatin, and ifosfamide. Functional validation of selected candidates, IER3 and S100A2, is also provided. Although the limitations of this approach are appreciated, it has produced detailed data that are practically impossible to obtain on osteosarcoma biopsies from chemotherapy-naive patients. Whereas osteosarcoma patients are treated with polychemotherapy, the results here presented relate candidate biomarkers to single-drug activities. The gene expression profiles contain several novel biomarker candidates that may be used to predict the responsiveness of osteosarcoma to doxorubicin, cisplatin, and ifosfamide. The validity and potential of selected candidates will be studied in clinical specimens obtained from osteosarcoma patients.

Chemotherapy has clearly improved the outcome for patients with osteosarcoma. For patients with resectable primary tumors and no overt metastatic disease at diagnosis, long-term survival rates of ∼70% can be achieved compared with the ∼20% cure rate previously obtained by surgery alone (1, 2). One of the most powerful prognostic indicators is the degree of necrosis in the primary tumor induced by preoperative chemotherapy (3, 4). Current treatment regimens usually involve neoadjuvant and adjuvant chemotherapy with high-dose methotrexate, doxorubicin, cisplatin, and, more recently, ifosfamide. However, a plateau for what may be achieved with current drugs with known activity in osteosarcoma has been reached (57). Moreover, for osteosarcoma patients treated with the current highly aggressive chemotherapy regimens, acute and long-term toxicity is a significant problem, and it is likely that some of the patients with micrometastatic disease could be cured with less aggressive regimens (8). Hence, biomarkers that could identify chemoresistant tumors already at the time of primary biopsy would potentially benefit substantially to the management of osteosarcoma. Microarray approaches have extensively been used in many tumor types in attempts to identify biomarkers for further clinical validation. In osteosarcoma, relatively few such studies have been reported, presumably reflecting that osteosarcoma is a relatively rare disease for which it is difficult to obtain sufficient tumor tissue from untreated patients. Recently, however, interesting results have been obtained on pretreatment biopsied samples and on tumor tissue obtained during surgery after preoperative chemotherapy (911). However, these studies cannot access genes related to individual drugs using patient's specimens. We have previously established a panel of osteosarcoma xenografts for the purpose of preclinical testing of novel drug candidates to treat osteosarcoma (12). Here, we report the results of microarray expression profiling of these xenografts to screen for potential markers for each of three of the drugs used in current clinical treatment protocols, resulting in three different subsets of genes that with high significance distinguished between poor and good responders to doxorubicin, cisplatin, and ifosfamide. The advantage of our approach permitted comparison of the effects of three commonly used drugs individually to the expression profile for each of them, which is impossible using clinical specimens.

Animals, xenografts, and assessment of chemotherapy response

All procedures involving animals were done according to protocols approved by the National Animal Research Authority and conducted according to the European Convention for the Protection of Vertebrates Used for Scientific Purposes. Female BALB/c nu/nu mice bred at the nude rodent facility at the Norwegian Radium Hospital were used. The animals were maintained, and experiments and assessment of chemotherapy response were conducted as described previously (12). In brief, the animals were 4 to 6 weeks old at the day of tumor implantation. Anesthesia was obtained with intraperitoneal injection of 0.1 mg/kg fentanyl, 5 mg/kg fluanisone, and 2.5 mg/kg midazolam. Tumor fragments (2 × 2 × 2 mm) were implanted subcutaneously in both flanks of nude mice. The animals were randomized for treatment according to tumor size when the average tumor diameters were ∼6 mm. Each experiment contained four subgroups of mice that were treated intravenously with saline (control), 8 mg/kg doxorubicin (Nycomed Pharma), 5 mg/kg cisplatin (Platinol; Bristol-Myers Squibb), or 240 mg/kg ifosfamide (Holoxan; Asta Medica). In addition, tumors were harvested for the purpose of microarray gene expression analysis from a fifth subgroup of animals on day 0. The antitumor activity of each drug was quantified calculating the specific growth delay (SGD) after one and two relative tumor volume doubling times (SGD200 or SGD400) and the maximal growth inhibition (T/C%).

Ten human osteosarcoma xenografts were used, eight of which have been described previously (12), and in addition, two newly established xenografts were included. These originated from a 32-year-old male with an extremity located fibroblastic osteosarcoma of malignancy grade 4 (GKAMX) and from a 13-year-old female with an extremity located osteoblastic osteosarcoma of malignancy grade 4 (CTPX).

Sample preparation, array hybridization, and gene expression analysis

Frozen xenografts were grinded by a stainless-steel mortar and pestle under liquid nitrogen. The tissue powder was then transferred to a conical tube and total RNA was isolated using the Trizol reagent (Invitrogen) followed by DNase treatment for 30 min at 37°C (RQ1 RNase-free DNase; Promega). CodeLink UniSet Human 20K Oligo Bioarray (GE Healthcare), containing ∼20,000 gene probes, was used to generate gene expression profiles. All reagents were provided in the CodeLink expression assay kit (GE Healthcare), except where noted. cRNA synthesis was done as per manufacturer's instructions. Using 2 μg total RNA, first-strand cDNA was generated by reverse transcriptase and a T7 primer. Subsequently, second-strand cDNA was produced using DNA polymerase I and RNase H. The resulting double-stranded cDNA was purified on a QIAquick column (Qiagen) and cRNA was generated via an in vitro transcription reaction using T7 RNA polymerase and biotin-11-UTP (Perkin-Elmer) at 37°C for 14 h. cRNA was purified on a RNeasy column (Qiagen) and quantified by UV spectrophotometry, and 10 μg biotin-labeled cRNA was then fragmented by heating at 94°C for 20 min in the presence of magnesium buffer. The fragmented cRNA was hybridized overnight at 37°C in hybridization buffer to a UniSet Human 20K Bioarray in a shaking incubator at 300 rpm. After hybridization, the arrays were washed in 0.75× TNT buffer [1× TNT: 0.1 mol/L Tris-HCl (pH 7.6), 0.15 mol/L NaCl, and 0.05% Tween 20] at 46°C for 1 h followed by incubation with Cy5-streptavadin (GE Healthcare) at room temperature for 30 min in the dark. Arrays were then washed in 1× TNT four times for 5 min each. The slides were then dried by centrifugation and kept in the dark until scanning. Images were captured on an Axon GenePix 4200A scanner. The resulting image was quantified and the intensity of each spot was divided by the median spot intensity to provide a scaled and comparable number across multiple arrays. After dot grid and QC, CodeLink software generates export files for analysis by GeneSpring software 7.2 (Agilent).

Assessment of total RNA and cRNA concentration, purity, and quality

The concentrations of both total RNA and cRNA were determined by measurement of absorbance at 260 nm (A260) using Nanodrop spectrophotometer. Only samples with A260:A280 ratio >1.8 were regarded as satisfactory pure. The quality of total RNA and cRNA was also evaluated by gel electrophoresis.

Quantitative reverse transcription-PCR

For real-time quantitative reverse transcription-PCR (qRT-PCR), the SYBR Green master mix from Applied Biosystems was used. qRT-PCR was done on an ABI 7500HT instrument under the following conditions: 25°C for 10 min and 37°C for 2 h for reverse transcription. For amplification, 5 min initial denaturation at 95°C for 40 cycles of 15 s denaturation and 1 min annealing extension at 37°C. The gene expression CT value from each sample was calculated by normalizing with internal housekeeping gene GAPDH and relative quantization values were plotted. qRT-PCR primers were purchased from Integrated DNA Technologies and Eurogentec (Belgium; S100A2). For the individual reactions, primers sequences listed in Supplementary Table S1 were used.

Cell culture and small interfering RNA gene silencing

The human osteosarcoma cell line OHS was routinely maintained in RPMI 1640 (BioWhittaker) supplemented with 10% fetal bovine serum (PAA Laboratories) and 2 mmol/L glutaMAX (Life Technologies). The following small interfering RNA (siRNA) sequences were purchased from Ambion: S100A2 (ID:17214) 5′-CAGGAAAACAGCAUACUCCtg-3′ and IER3 (ID:6661) 5′-AGCAGCAGAAAGAGAAGAAGCCtt-3′. Silencer Negative Control #1 siRNA was used as a scrambled control. The effects of gene silencing with siRNA on sensitivity to 4-hydroperoxyifosfamide (D-18851; Universität Bielefeld) were assessed by the clonogenic assay. Briefly, OHS cells were seeded in 6-well plates (Costar 3516; Corning) and incubated overnight to achieve a confluency of 30% to 50%, before transfections with siRNA, using the Lipofectamine 2000 (Invitrogen) reagent according to the manufacturer's protocol, using 1 μg/mL Lipofectamine 2000 and 30 nmol/L siRNA. After transfections, the cells were incubated for 24 h and trypsinized and counted. Known numbers were then replated in triplicate wells and incubated for 24 h, allowing the cells to attach before treatment with indicated concentrations of 4-hydroperoxyifosfamide and finally incubated for 7 days to allow macroscopic colony development. Colonies were counted and the plating efficiency and surviving fraction for given treatments were calculated based on the survival of cells treated with 4-hydroperoxyifosfamide compared with vehicle-treated controls. 4-Hydroperoxyifosfamide was dissolved in water immediately before use to obtain stock concentrations that were added to the growth medium at a 1:30 ratio.

Western blot

Preparation of whole-cell lysates was done by addition of lysis buffer [150 mmol/L NaCl, 50 mmol/L Tris-HCl (pH 7.5), 0.1% NP-40, 10 μg/mL of each leupeptin hemisulfate, aprotinin, and pepstatin A, 20 mmol/L β-glycerophosphate, 1 mmol/L phenylmethylsulfonyl fluoride, 1 mmol/L sodium orthovanadate, and 100 mmol/L sodium fluoride] to dry cell pellets, left on ice for 15 min, sonicated, and frozen at −80°C. Upon use, the samples were centrifuged to remove cell debris. Total protein lysate from each sample (100 μg) was separated on 8% to 12% SDS-PAGE and transferred onto Immobilon-P membranes (Millipore) according to the manufacturer's manual. After blockage of nonspecific binding sites with 10% nonfat dry milk in TBS with 0.25% Tween, blots were incubated for 1 h at room temperature with a mouse monoclonal anti-S100A2 (clone SH-L1; Sigma), a rabbit polyclonal anti-IER3 (Abcam), or a mouse monoclonal anti-α-tubulin antibody (0.3 μg/mL; Oncogene Research Products). After washing, the blots were incubated for 1 h at room temperature with a rabbit anti-mouse or a goat anti-rabbit secondary antibody (DAKO) diluted 1:4,000. Signals were visualized using the enhanced chemiluminescence substrate (Amersham Pharmacia Biotech).

Statistical analysis

GeneSpring version 7.2 was used for normalization, filtering, statistical and Gene Ontology (GO) analysis of the microarray data, and hierarchical clustering of the selected genes. The expression data for each of the hybridizations were first normalized to their global median. Then, the expression of each gene was normalized to its median. Before the statistical analysis, genes flagged present in less than half of the conditions and that had an average expression signal below 200 in at least one of the groups were filtered out of the data sets. To identify genes with a high differential expression among the two groups analyzed, a one-way ANOVA test with a Benjamini and Hochberg multiple test correction allowing for a given false discovery rate of was done. Hierarchical clustering was done using the standard correlation option of the GeneSpring clustering tool. Finally, the integrated GeneSpring GO browser was used for category enrichment analysis.

Response of human osteosarcoma xenografts to doxorubicin, cisplatin, and ifosfamide

Table 1 shows the classification of human osteosarcoma xenografts according to their response to doxorubicin, cisplatin, and ifosfamide. For analysis of the microarray expression data, we grouped the xenografts as good (++/+++/++++) or poor (−/+) responders. Four of the xenografts responded well doxorubicin, three to cisplatin, and five to ifosfamide. The sensitivity profiles for doxorubicin and cisplatin were similar with only one cisplatin-resistant xenograft responding well to doxorubicin. The sensitivity profile for ifosfamide was somewhat different from the two other drugs, with overall five responding xenografts, but two of the xenografts that responded poorly were sensitive to doxorubicin and cisplatin.

Table 1.

Response of human osteosarcoma xenografts to doxorubicin, cisplatin, and ifosfamide

XenograftTDDoxorubicinCisplatinIfosfamide
SGD200SGD400T/C%EfficacyGrouping-expression analysis (response)SGD200SGD400T/C%EfficacyGrouping-expression analysis (response)SGD200SGD400T/C%EfficacyGrouping-expression analysis (response)
OHS 0.4 0.4 62  Poor 0.3 0.6 49  Poor >37.6 >14.2 ++++ Good 
CTPX 2.9 2.9 10 +++ Good 1.6 1.1 25 ++ Good >4.3 >4.3 ++++ Good 
SBX 6.5 0.8 1.7 28 ++ Good 75  Poor >22.1 >9.3 ++++ Good 
ALSKX 11 0.7 52  Poor 0.1 0.2 59  Poor 3.2 1.8 14 +++ Good 
AOX 10 0.5 58  Poor 0.5 0.3 69  Poor 3.7 1.9 12 +++ Good 
TTX 4.8 1.7 17 +++ Good 7.6 2.7 ++++ Good 1.8 0.7 50 Poor 
GKAMX 0.9 1.5 25 ++ Good 1.5 35 ++ Good 1.3 1.1 28 Poor 
TPX 13 0.1 0.1 83  Poor 0.2 0.1 72  Poor 1.3 0.7 36 Poor 
TSX pr1 4.5 0.3 0.2 71  Poor 0.6 0.4 74  Poor 0.8 0.4 57  Poor 
KPDX 0.9 0.8 47  Poor 0.2 86  Poor 0.5 68  Poor 
XenograftTDDoxorubicinCisplatinIfosfamide
SGD200SGD400T/C%EfficacyGrouping-expression analysis (response)SGD200SGD400T/C%EfficacyGrouping-expression analysis (response)SGD200SGD400T/C%EfficacyGrouping-expression analysis (response)
OHS 0.4 0.4 62  Poor 0.3 0.6 49  Poor >37.6 >14.2 ++++ Good 
CTPX 2.9 2.9 10 +++ Good 1.6 1.1 25 ++ Good >4.3 >4.3 ++++ Good 
SBX 6.5 0.8 1.7 28 ++ Good 75  Poor >22.1 >9.3 ++++ Good 
ALSKX 11 0.7 52  Poor 0.1 0.2 59  Poor 3.2 1.8 14 +++ Good 
AOX 10 0.5 58  Poor 0.5 0.3 69  Poor 3.7 1.9 12 +++ Good 
TTX 4.8 1.7 17 +++ Good 7.6 2.7 ++++ Good 1.8 0.7 50 Poor 
GKAMX 0.9 1.5 25 ++ Good 1.5 35 ++ Good 1.3 1.1 28 Poor 
TPX 13 0.1 0.1 83  Poor 0.2 0.1 72  Poor 1.3 0.7 36 Poor 
TSX pr1 4.5 0.3 0.2 71  Poor 0.6 0.4 74  Poor 0.8 0.4 57  Poor 
KPDX 0.9 0.8 47  Poor 0.2 86  Poor 0.5 68  Poor 

Abbreviations: TD. tumor doubling time; T/C%. maximal growth inhibition.

Differentially expressed genes

Expression profiles established from the 10 human osteosarcoma xenografts were based on mean values from triplicate hybridizations. When filtering was done as described in Materials and Methods, ∼3,600 genes were found eligible for statistical analysis. With a false discovery rate of 5%, 85 genes were identified as differentially expressed between good and poor responders to doxorubicin. Of these, 48 were expressed higher and 37 lower in the poorly responsive xenografts compared with those that responded well (Fig. 1A). Seventy-four genes were identified as differentially expressed between cisplatin poor and good responders (false discovery rate = 2%), 38 of which were expressed higher and 36 were lower in the poorly responsive group (Fig. 1B). For ifosfamide, 118 genes were identified as differentially expressed between poor and good responders (false discovery rate = 0.5%), and 67 and 51 of these were higher and lower expressed in the poorly responsive xenografts, respectively (Fig. 1C). Lists for all three expression profiles including full gene names and accession numbers are shown in Supplementary Table S2A to C. For all three expression profiles, clustering of significant genes resulted in two distinct groups in which poor or good responder xenografts clustered together. Among the genes that showed significant differential expression between xenografts that responded well or poorly to cisplatin, 21 were overlapping with the comparable gene list for doxorubicin. In contrast, only 2 genes were common in the expression profiles for cisplatin and ifosfamide and none when comparing the profiles for doxorubicin and ifosfamide.

Fig. 1.

Heat maps for genes that classify human osteosarcoma xenografts according to chemosensitivity. Levels of expression represent mean values from triplicate array hybridization and are presented as below (green) or above (red) the median expression of each gene (black). Blue, genes that were flagged absent in the representing arrays. These lists of genes were selected by a one-way ANOVA analysis with a Benjamini-Hochberg multiple test correction allowing false discovery rates of 5% for doxorubicin (A; 85 genes), 2% for cisplatin (B; 74 genes), and 0.5% for ifosfamide (C; 118 genes). GenBank accessions and full gene names are listed in Supplementary Table S2A to C.

Fig. 1.

Heat maps for genes that classify human osteosarcoma xenografts according to chemosensitivity. Levels of expression represent mean values from triplicate array hybridization and are presented as below (green) or above (red) the median expression of each gene (black). Blue, genes that were flagged absent in the representing arrays. These lists of genes were selected by a one-way ANOVA analysis with a Benjamini-Hochberg multiple test correction allowing false discovery rates of 5% for doxorubicin (A; 85 genes), 2% for cisplatin (B; 74 genes), and 0.5% for ifosfamide (C; 118 genes). GenBank accessions and full gene names are listed in Supplementary Table S2A to C.

Close modal

qRT-PCR validation of selected genes

Ten of the genes identified as differentially expressed in poor or good responding osteosarcoma xenografts to doxorubicin, cisplatin, or ifosfamide were selected for validation of the microarray data by qRT-PCR: FABP3 and TGFBI (doxorubicin); DPP7, NCALD, and SSX1 (doxorubicin and cisplatin); and IER3, GADD45A, UBE2L6, EPB41L3, and ALPL (Ifosfamide). The expression levels for these genes assessed by qRT-PCR confirmed the microarray data, illustrated as fold differences between poor and good responders for selected genes for each drug (Table 2). Moreover, there were significant correlations between results obtained by the two methods for all 10 genes (r = 0.71-0.98; P ≤ 0.02).

Table 2.

Fold difference in expression levels of selected genes differentially expressed in poor and good responding xenografts to doxorubicin, cisplatin, or ifosfamide

GeneFold difference (poor responders/good responders)
MicroarrayqRT-PCR
Doxorubicin 
    FABP3 2.8 3.7 
    NCALD −3 −5.7 
    TGFBI −13.8 −13.3 
    SSX1 −16.6 −3336 
Cisplatin 
    DDP7 3.7 7.1 
    NCALD −5.5 −8.9 
    SSX1 −4.7 −14.2 
Ifosfamide 
    IER3 11.7 19.3 
    UBE2L6 10.5 9.3 
    GADD45A 4.3 11.9 
    ALPL −40.8 −36 
    EPB41L3 −52.4 −176 
GeneFold difference (poor responders/good responders)
MicroarrayqRT-PCR
Doxorubicin 
    FABP3 2.8 3.7 
    NCALD −3 −5.7 
    TGFBI −13.8 −13.3 
    SSX1 −16.6 −3336 
Cisplatin 
    DDP7 3.7 7.1 
    NCALD −5.5 −8.9 
    SSX1 −4.7 −14.2 
Ifosfamide 
    IER3 11.7 19.3 
    UBE2L6 10.5 9.3 
    GADD45A 4.3 11.9 
    ALPL −40.8 −36 
    EPB41L3 −52.4 −176 

NOTE: Comparison of microarray and qRT-PCR results.

GO categories enriched of differentially expressed genes

GO categories that were enriched with a significance level of P < 0.05 for genes with either significantly higher or lower expression in poorly compared with good responding xenografts are shown in Supplementary Table S3. Categories containing altogether five or more genes are included. For xenografts responding poorly to doxorubicin, selective enrichment of genes with significantly higher expression in poor responders was found in ontology categories related to energy metabolism and mitochondrion and the ontology category response to abiotic stimulus. In contrast, extracellular region was the only the category that, with convincing selectivity, was significantly enriched by genes with low expression in doxorubicin poor responders. This category constituted genes that also received the annotations extracellular matrix (n = 4) and/or skeletal development (n = 3). For cisplatin, categories selectively enriched by genes upregulated in poorly responding xenografts to cisplatin were RNA metabolism, hydrolase activity, cytoplasm, and endoplasmic reticulum. As for doxorubicin genes with mitochondrial annotation were upregulated in poor responder xenografts, although the enrichment scores did not reach significance (n = 4; P = 0.11). For ifosfamide, genes with high expression in poor responders were most strongly overrepresented in the category of negative regulation of physiologic process, apoptosis, calcium ion binding, and microtubule cytoskeleton. The GO categories most significantly enriched by genes with lower expression in poorly responding xenografts were skeletal and organ development. Many of the same genes were also found in two other significant categories: regulation of transcription and extracellular region.

Functional validation of selected genes

To investigate whether any of the identified genes could be functionally related to chemotherapy resistance, siRNA-mediated gene silencing of two genes that were significantly higher expressed in xenografts resistant to ifosfamide, S100A2 and IER3, were done in the human osteosarcoma OHS cell line. Effects on sensitivity to 4-hydroperoxyifosfamide, which is the active metabolite of ifosfamide, were then assessed by the clonogenic assay. siRNA reduced transcript levels of S100A2 to ∼5% of the levels in control untransfected OHS cells as assessed by qRT-PCR 24 h after transfections (Fig. 2A). At the protein level, the expression of S100A2 was reduced 48 h after transfection and almost abolished after 96 h (Fig. 2A). For IER3, transcript levels were only reduced ∼50% by the transfected siRNA and a transient reduction in protein level was seen after 48 h (Fig. 2B). For IER3, the prominent bands detected by Western immunoblot analysis correspond to a nonglycosylated form of 17 kDa and a glycosylated form of 26 to 29 kDa, previously published by others (13, 14). Gene silencing with siRNA to IER3 but not S100A2 or a scrambled negative control significantly reduced colony formation of OHS cells compared with untransfected cells (Fig. 2C). The effects of gene silencing on the sensitivity of OHS cells to 4-hydroperoxyifosfamide are shown in Fig. 2D. For OHS cells transfected with siRNAs to either S100A2 or IER3 (IC50 ∼0.3 μmol/L), significant leftward shifts of the dose-response curves for 4-hydroperoxyifosfamide, compared with both untransfected cells and the negative control (IC50 ∼0.6 μmol/L), were observed.

Fig. 2.

Effects of gene silencing with siRNAs to S100A2 (siS100A2) or IER3 (siIER3) compared with a scrambled negative control (siNEG) and untransfected cells in the OHS human osteosarcoma cell line. A, left, transcriptional levels of S100A2 24 h after siRNA transfections expressed relative to untransfected cells (bars, SD, duplicates); Right, Western blot showing protein levels of S100A2 after siRNA-mediated transcript knockdown. Recombinant His-tagged S100A2 was used as a positive control. B, left, transcriptional levels of IER3 24 h after siRNA transfections expressed relative to untransfected cells (bars, SD, duplicates); right, Western blot showing protein levels of IER3 after siRNA-mediated transcript knockdown. Two dominant bands are observed, a nonglycosylated form of ∼17 kDa and a glycosylated form of ∼26 kDa. C, silencing of IER3 but not S100A2 or a scrambled negative control reduces colony formation of OHS cells (bars, SE, 3 experiments). D, silencing of IER3 and S100A2 sensitizes OHS cells to 4-hydroperoxyifosfamide (bars, SE, 3 experiments).

Fig. 2.

Effects of gene silencing with siRNAs to S100A2 (siS100A2) or IER3 (siIER3) compared with a scrambled negative control (siNEG) and untransfected cells in the OHS human osteosarcoma cell line. A, left, transcriptional levels of S100A2 24 h after siRNA transfections expressed relative to untransfected cells (bars, SD, duplicates); Right, Western blot showing protein levels of S100A2 after siRNA-mediated transcript knockdown. Recombinant His-tagged S100A2 was used as a positive control. B, left, transcriptional levels of IER3 24 h after siRNA transfections expressed relative to untransfected cells (bars, SD, duplicates); right, Western blot showing protein levels of IER3 after siRNA-mediated transcript knockdown. Two dominant bands are observed, a nonglycosylated form of ∼17 kDa and a glycosylated form of ∼26 kDa. C, silencing of IER3 but not S100A2 or a scrambled negative control reduces colony formation of OHS cells (bars, SE, 3 experiments). D, silencing of IER3 and S100A2 sensitizes OHS cells to 4-hydroperoxyifosfamide (bars, SE, 3 experiments).

Close modal

In osteosarcoma, the introduction and optimization of aggressive preoperative and postoperative chemotherapy has decisively improved patient survival. However, a significant number of patients have tumors resistant to the drugs routinely used and hence receive a tough and toxic treatment without any therapeutic benefit. There is an obvious need for markers that predict tumor response and resistance, and gene expression profiling might provide promising leads that can be validated for their clinical potential. One key obstacle for performing such studies on clinical specimens is the difficulty in obtaining pretreatment osteosarcoma samples in an amount and quality needed for such studies, and because the patients receive multidrug chemotherapy, genes involved in resistance to the individual drugs cannot readily be identified. Our approach of using osteosarcoma xenografts with different sensitivities to clinically used drugs circumvents these problems. Outliers were selected with low false discovery rates; expression assessed with qRT-PCR correlated tightly with the array data.

We are aware of the fact that even a large panel of 10 xenografts may not cover the range of biological characteristics of clinical osteosarcoma. Moreover, from a statistical point of view, it is a relatively low number of tumors, and the reproducibility of results in xenografts with low or intermediate sensitivity is therefore of concern. Thus, the selection of genes that correlate with response to chemotherapy could change markedly if only one xenograft moved from one response category to the other. Future studies of this type should therefore be conducted in xenografts with either very high or very low sensitivity to the drug studied as was the case here for ifosfamide. However, we still feel that our xenograft panel represents a valuable tool for identifying new prognostic markers, and the present results support this conclusion. First, some of the identified genes have been shown previously to correlate with prognosis or chemotherapy response in osteosarcoma. Secondly, GO-guided analysis of the gene lists obtained identified genes and groups of genes that can be associated with known functions related to mechanisms of drug sensitivity and resistance. Third, gene silencing of two of the identified genes with significantly enhanced expression in ifosfamide poor responder xenografts, IER3 and S100A2, sensitized human osteosarcoma OHS cells to treatment with 4-hydroperoxyifosfamide.

Previously, osteosarcoma gene expression profiles that characterize poor or good responder osteosarcomas using clinical specimens from patients have been reported. Some of the genes identified in these studies were also found in our lists, such as MAGED1 (9), IER3, PAI1, and AXNA2 (11), and HSP70 and MCM2 (10). Moreover, HSP27 and HSP70, which was associated with poor response to doxorubicin and cisplatin in our xenograft panel, have been shown previously to correlate with poor response to neoadjuvant chemotherapy when assessed by immunohistochemistry (1517). In the previous studies, the combined effect of several drugs used in preoperative regimens was assessed by the degree of tumor necrosis at the time of surgery. The advantage of our approach permitted comparison of the effects of three commonly used drugs individually to the expression profile for each of them, which is impossible using clinical specimens. The advantage was clearly shown here in that it gives a better insight in the molecular mechanisms underlying resistance to the individual drugs, thereby providing the possibility to identify leads to genes and pathways that may be targeted therapeutically in addition to detect markers of putative prognostic value.

GO enrichment analysis was done to identify possible groups of genes associated to the chemoresistant phenotype. For doxorubicin, the most striking finding was that mitochondrial location and ontology categories related to energy metabolism were selectively enriched by genes with higher expression levels in xenografts responding poorly. This finding may link the lack of doxorubicin response to mitochondrial malfunctions or tumor hypoxia, phenomena with a well-established association to drug resistance (1821). Moreover, the GO category response to abiotic stimulus contains genes that may interplay to inhibit mitochondrial/intrinsic apoptotic pathways, such as the heat shock proteins HSP27 and HSP70 (22), Bcl-2–associated athanogene 2 (23, 24), and SRI (Sorcin). Sorcin predicts poor outcome and response to chemotherapy in acute myeloid leukemia and may therefore be of particular interest (25, 26). Conversely, the category extracellular region was significantly and selectively enriched by genes downregulated. These genes included COL12A1, INHBA, and MGP, which are involved in skeletal development, and TGFBI, which is a cell adhesion molecule that can inhibit differentiation and promote apoptosis and chemosensitivity (2729).

Genes with high expression in cisplatin poor responders were enriched by genes annotated to the endoplasmatic reticulum. One of these, RTN3, is an endoplasmic reticulum stress response gene that has been reported to facilitate the antiapoptotic activity of Bcl-2 (30). Further, two proapoptotic genes, GALC and PHLDA1, were strongly downregulated. GALC causes cellular accumulation of ceramide, which is an important second messenger of apoptosis (31), whereas downregulation of PHLDA1 contribute to apoptotic resistance in malignant melanoma (32). Interesting genes that overlapped with the profile for doxorubicin include HSP70 as well as DPP7, which has been described as an important regulator of apoptotic threshold in resting lymphocytes (33).

For ifosfamide, genes involved in apoptosis regulation were only identified in the group of genes that had higher expression in xenografts that responded poorly (AXNA4, Aven, IER3, GADD45A, and SH3KBP1). In addition, at least two other highly deregulated genes, FHL2 and MLF1, can promote survival through transcriptional regulation (3437). This suggests that antiapoptotic mechanisms might be involved in mediating resistance to ifosfamide. Taken together, genes involved in development and transcriptional regulation and annotated to the category extracellular region indicate that poorly responding xenografts are characterized by dedifferentiation, tumor aggressiveness, and invasiveness. Genes with lower expression are involved in transforming growth factor-β superfamily signaling and osteoblastic and bone differentiation, including BMP4, MADH6, and ID3 (transforming growth factor-β pathway), MSX2 (38), MEF2C (39), COL11A2 (40), and ALPL. Conversely, genes with higher expression include the two transcription regulators CUTL1 and TCF8 (41, 42) and three proteolytic factors PLAU(uPA), SERPINE1 (PAI-1), and CTSL (cathepsin L), associated with tumor invasion and metastasis.

To further validate our findings, we performed siRNA-mediated gene silencing on two selected genes, differentially expressed between ifosfamide poor and good responder xenografts, IER3 and S100A2. Knockdown of each of these genes gave a modest but significant degree of chemosensitization. This is likely to reflect the notion that a network of genes, rather than a single gene, drives the chemoresistant phenotype. IER3, initially characterized as a radiation-inducible early response gene (43), exerts positive or negative regulation of apoptosis depending on the cell type (14, 4449). We have shown that IER3 correlated inversely with response to ifosfamide in osteosarcoma xenografts and that a partial and transient silencing of IER3 sensitized osteosarcoma cells to ifosfamide and significantly impaired the ability of OHS cells to form colonies in vitro. Seemingly in contrast to this, the clinical study by Mintz et al. (11) showed that IER3 was overexpressed in biopsies from tumors with good histologic response. Because the preoperative chemotherapy in that study consisted of methotrexate, doxorubicin, and cisplatin, the findings are not necessarily contradictory to ours, as the effects of different drugs were studied.

In summary, we have obtained gene expression profiles from a panel of osteosarcoma xenografts that in parallel were tested for their sensitivity to doxorubicin, cisplatin, and ifosfamide. Although the limitations of this approach are appreciated, it has produced detailed data that are practically impossible to obtain on osteosarcoma biopsies from chemotherapy-naive patients. The xenografts showed a similar response to doxorubicin and cisplatin, whereas their chemosensitivity profile to ifosfamide was somewhat different. Despite these differences, the common features of the poorly responsive phenotype seem to be a drive toward dedifferentiation, tumor aggressiveness, and resistance to undergo programmed cell death. Moreover, we have shown that silencing of the S100A2 and IER3 genes sensitize osteosarcoma cells to ifosfamide. The gene expression profiles here presented contain several novel biomarker candidates that may be used to predict the responsiveness of osteosarcoma to doxorubicin, cisplatin, and ifosfamide. The validity and potential of selected candidates will be studied in clinical specimens obtained from osteosarcoma patients.

No potential conflicts of interest were disclosed.

We thank Gunhild M. Maelandsmo for helpful advice and Tove Øyjord, Thomas Halvorsen, and Marita Martinsen for excellent technical assistance.

1
Souhami
R
,
Cannon
S
. 
Osteosarcoma
. In:
Peckham
M
,
Pinedo
HM
,
Veronesi
U
, editors.
Oxford textbook of oncology
.
Oxford
:
Oxford University Press
; 
1995
, p.
1969
76
.
2
Malawer
MM
,
Link
MP
,
Donaldson
SS
. 
Sarcomas of the bone
. In:
DeVita
VT
,
Hellmann
S
,
Rosenberg
SA
, editors.
Cancer: principles and practice of oncology
. 6th ed.
2001
, p.
1891
936
.
3
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
.
4
Provisor
AJ
,
Ettinger
LJ
,
Nachman
JB
, et al
. 
Treatment of nonmetastatic osteosarcoma of the extremity with preoperative and postoperative chemotherapy: a report from the Children's Cancer Group
.
J Clin Oncol
1997
;
15
:
76
84
.
5
Bacci
G
,
Briccoli
A
,
Rocca
M
, et al
. 
Neoadjuvant chemotherapy for osteosarcoma of the extremities with metastases at presentation: recent experience at the Rizzoli Institute in 57 patients treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide
.
Ann Oncol
2003
;
14
:
1126
34
.
6
Gorlick
R
,
Meyers
PA
. 
Osteosarcoma necrosis following chemotherapy: innate biology versus treatment-specific
.
J Pediatr Hematol Oncol
2003
;
25
:
840
1
.
7
Smeland
S
,
Muller
C
,
Alvegard
TA
, et al
. 
Scandinavian Sarcoma Group Osteosarcoma Study SSG VIII: prognostic factors for outcome and the role of replacement salvage chemotherapy for poor histological responders
.
Eur J Cancer
2003
;
39
:
488
94
.
8
Bruland
OS
,
Pihl
A
. 
On the current management of osteosarcoma. A critical evaluation and a proposal for a modified treatment strategy
.
Eur J Cancer
1997
;
33
:
1725
31
.
9
Ochi
K
,
Daigo
Y
,
Katagiri
T
, et al
. 
Prediction of response to neoadjuvant chemotherapy for osteosarcoma by gene-expression profiles
.
Int J Oncol
2004
;
24
:
647
55
.
10
Man
T-K
,
Chintagumpala
M
,
Visvanathan
J
, et al
. 
Expression profiles of osteosarcoma that can predict response to chemotherapy 10.1158/0008-5472.CAN-05-0985
.
Cancer Res
2005
;
65
:
8142
50
.
11
Mintz
MB
,
Sowers
R
,
Brown
KM
, et al
. 
An expression signature classifies chemotherapy-resistant pediatric osteosarcoma 10.1158/0008-5472.CAN-04-2463
.
Cancer Res
2005
;
65
:
1748
54
.
12
Bruheim
S
,
Bruland
OS
,
Breistol
K
,
Maelandsmo
GM
,
Fodstad
O
. 
Human osteosarcoma xenografts and their sensitivity to chemotherapy
.
Pathol Oncol Res
2004
;
10
:
133
41
.
13
Feldmann
KA
,
Pittelkow
MR
,
Roche
PC
,
Kumar
R
,
Grande
JP
. 
Expression of an immediate early gene, IEX-1, in human tissues
.
Histochem Cell Biol
2001
;
115
:
489
97
.
14
Shen
L
,
Guo
J
,
Santos-Berrios
C
,
Wu
MX
. 
Distinct domains for anti- and pro-apoptotic activities of IEX-1
.
J Biol Chem
2006
;
281
:
15304
11
.
15
Uozaki
H
,
Horiuchi
H
,
Ishida
T
,
Iijima
T
,
Imamura
T
,
Machinami
R
. 
Overexpression of resistance-related proteins (metallothioneins, glutathione-S-transferase π, heat shock protein 27, and lung resistance-related protein) in osteosarcoma. Relationship with poor prognosis
.
Cancer
1997
;
79
:
2336
44
.
16
Trieb
K
,
Lechleitner
T
,
Lang
S
,
Windhager
R
,
Kotz
R
,
Dirnhofer
S
. 
Heat shock protein 72 expression in osteosarcomas correlates with good response to neoadjuvant chemotherapy
.
Hum Pathol
1998
;
29
:
1050
5
.
17
Uozaki
H
,
Ishida
T
,
Kakiuchi
C
, et al
. 
Expression of heat shock proteins in osteosarcoma and its relationship to prognosis
.
Pathol Res Pract
2000
;
196
:
665
73
.
18
Carew
JS
,
Huang
P
. 
Mitochondrial defects in cancer
.
Mol Cancer
2002
;
1
:
9
.
19
Xu
RH
,
Pelicano
H
,
Zhou
Y
, et al
. 
Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia
.
Cancer Res
2005
;
65
:
613
21
.
20
Salvioli
S
,
Storci
G
,
Pinti
M
, et al
. 
Apoptosis-resistant phenotype in HL-60-derived cells HCW-2 is related to changes in expression of stress-induced proteins that impact on redox status and mitochondrial metabolism
.
Cell Death Differ
2003
;
10
:
163
74
.
21
Harper
ME
,
Antoniou
A
,
Villalobos-Menuey
E
, et al
. 
Characterization of a novel metabolic strategy used by drug-resistant tumor cells
.
FASEB J
2002
;
16
:
1550
7
.
22
Garrido
C
,
Brunet
M
,
Didelot
C
,
Zermati
Y
,
Schmitt
E
,
Kroemer
G
. 
Heat shock proteins 27 and 70: anti-apoptotic proteins with tumorigenic properties
.
Cell Cycle
2006
;
5
:
2592
601
.
23
Ueda
K
,
Kosako
H
,
Fukui
Y
,
Hattori
S
. 
Proteomic identification of Bcl2-associated athanogene 2 as a novel MAPK-activated protein kinase 2 substrate
.
J Biol Chem
2004
;
279
:
41815
21
.
24
Dai
Q
,
Qian
SB
,
Li
HH
, et al
. 
Regulation of the cytoplasmic quality control protein degradation pathway by BAG2
.
J Biol Chem
2005
;
280
:
38673
81
.
25
Tan
Y
,
Li
G
,
Zhao
C
, et al
. 
Expression of sorcin predicts poor outcome in acute myeloid leukemia
.
Leuk Res
2003
;
27
:
125
31
.
26
Qi
J
,
Liu
N
,
Zhou
Y
, et al
. 
Overexpression of sorcin in multidrug resistant human leukemia cells and its role in regulating cell apoptosis
.
Biochem Biophys Res Commun
2006
;
349
:
303
9
.
27
Kim
JE
,
Kim
SJ
,
Jeong
HW
, et al
. 
RGD peptides released from β ig-h3, a TGF-β-induced cell-adhesive molecule, mediate apoptosis
.
Oncogene
2003
;
22
:
2045
53
.
28
Thapa
N
,
Kang
KB
,
Kim
IS
. 
β ig-h3 mediates osteoblast adhesion and inhibits differentiation
.
Bone
2005
;
36
:
232
42
.
29
Ahmed
AA
,
Mills
AD
,
Ibrahim
AE
, et al
. 
The extracellular matrix protein TGFBI induces microtubule stabilization and sensitizes ovarian cancers to paclitaxel
.
Cancer Cell
2007
;
12
:
514
27
.
30
Wan
Q
,
Kuang
E
,
Dong
W
, et al
. 
Reticulon 3 mediates Bcl-2 accumulation in mitochondria in response to endoplasmic reticulum stress
.
Apoptosis
2007
;
12
:
319
28
.
31
Beier
UH
,
Gorogh
T
. 
Implications of galactocerebrosidase and galactosylcerebroside metabolism in cancer cells
.
Int J Cancer
2005
;
115
:
6
10
.
32
Neef
R
,
Kuske
MA
,
Prols
E
,
Johnson
JP
. 
Identification of the human PHLDA1/TDAG51 gene: down-regulation in metastatic melanoma contributes to apoptosis resistance and growth deregulation
.
Cancer Res
2002
;
62
:
5920
9
.
33
Chiravuri
M
,
Schmitz
T
,
Yardley
K
,
Underwood
R
,
Dayal
Y
,
Huber
BT
. 
A novel apoptotic pathway in quiescent lymphocytes identified by inhibition of a post-proline cleaving aminodipeptidase: a candidate target protease, quiescent cell proline dipeptidase
.
J Immunol
1999
;
163
:
3092
9
.
34
Lee
SW
,
Kim
EJ
,
Um
SJ
. 
FHL2 mediates p53-induced transcriptional activation through a direct association with HIPK2
.
Biochem Biophys Res Commun
2006
;
339
:
1056
62
.
35
Stilo
R
,
Leonardi
A
,
Formisano
L
,
Di Jeso
B
,
Vito
P
,
Liguoro
D
. 
TUCAN/CARDINAL and DRAL participate in a common pathway for modulation of NF-κB activation
.
FEBS Lett
2002
;
521
:
165
9
.
36
Lim
R
,
Winteringham
LN
,
Williams
JH
, et al
. 
MADM, a novel adaptor protein that mediates phosphorylation of the 14-3-3 binding site of myeloid leukemia factor 1
.
J Biol Chem
2002
;
277
:
40997
1008
.
37
Yoneda-Kato
N
,
Fukuhara
S
,
Kato
J
. 
Apoptosis induced by the myelodysplastic syndrome-associated NPM-MLF1 chimeric protein
.
Oncogene
1999
;
18
:
3716
24
.
38
Ma
L
,
Golden
S
,
Wu
L
,
Maxson
R
. 
The molecular basis of Boston-type craniosynostosis: the Pro148->His mutation in the N-terminal arm of the MSX2 homeodomain stabilizes DNA binding without altering nucleotide sequence preferences
. 
1996
:
1915
20
.
39
Mackie
EJ
,
Ahmed
YA
,
Tatarczuch
L
,
Chen
KS
,
Mirams
M
. 
Endochondral ossification: how cartilage is converted into bone in the developing skeleton
.
Int J Biochem Cell Biol
2008
;
40
:
46
62
.
40
Vikkula
M
,
Mariman
EC
,
Lui
VC
, et al
. 
Autosomal dominant and recessive osteochondrodysplasias associated with the COL11A2 locus
.
Cell
1995
;
80
:
431
7
.
41
Michl
P
,
Ramjaun
AR
,
Pardo
OE
, et al
. 
CUTL1 is a target of TGF(β) signaling that enhances cancer cell motility and invasiveness
.
Cancer Cell
2005
;
7
:
521
32
.
42
Reppe
S
,
Olstad
OK
,
Rian
E
,
Gautvik
VT
,
Gautvik
KM
,
Jemtland
R
. 
Butyrate response factor 1 is regulated by parathyroid hormone and bone morphogenetic protein-2 in osteoblastic cells
.
Biochem Biophys Res Commun
2004
;
324
:
218
23
.
43
Kondratyev
AD
,
Chung
KN
,
Jung
MO
. 
Identification and characterization of a radiation-inducible glycosylated human early-response gene
.
Cancer Res
1996
;
56
:
1498
502
.
44
Wu
MX
. 
Roles of the stress-induced gene IEX-1 in regulation of cell death and oncogenesis
.
Apoptosis
2003
;
8
:
11
8
.
45
Sebens Muerkoster
S
,
Rausch
AV
,
Isberner
A
, et al
. 
The apoptosis-inducing effect of gastrin on colorectal cancer cells relates to an increased IEX-1 expression mediating NF-κB inhibition
.
Oncogene
2008
;
27
:
1122
34
.
46
Arlt
A
,
Grobe
O
,
Sieke
A
, et al
. 
Expression of the NF-κB target gene IEX-1 (p22/PRG1) does not prevent cell death but instead triggers apoptosis in HeLa cells
.
Oncogene
2001
;
20
:
69
76
.
47
Grobe
O
,
Arlt
A
,
Ungefroren
H
, et al
. 
Functional disruption of IEX-1 expression by concatemeric hammerhead ribozymes alters growth properties of 293 cells
.
FEBS Lett
2001
;
494
:
196
200
.
48
Schilling
D
,
Pittelkow
MR
,
Kumar
R
. 
IEX-1, an immediate early gene, increases the rate of apoptosis in keratinocytes
.
Oncogene
2001
;
20
:
7992
7
.
49
Zhang
Y
,
Schlossman
SF
,
Edwards
RA
,
Ou
CN
,
Gu
J
,
Wu
MX
. 
Impaired apoptosis, extended duration of immune responses, and a lupus-like autoimmune disease in IEX-1-transgenic mice
.
Proc Natl Acad Sci U S A
2002
;
99
:
878
83
.

Competing Interests

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

Supplementary data