Gliomas are highly lethal neoplasms that cannot be cured by currently available therapies. Temozolomide is a recently introduced alkylating agent that has yielded a significant benefit in the treatment of high-grade gliomas. However, either de novo or acquired chemoresistance occurs frequently and has been attributed to increased levels of O6-methylguanine-DNA methyltransferase or to the loss of mismatch repair capacity. However, very few gliomas overexpress O6-methylguanine-DNA methyltransferase or are mismatch repair–deficient, suggesting that other mechanisms may be involved in the resistance to temozolomide. The purpose of the present study was to generate temozolomide-resistant variants from a human glioma cell line (SNB-19) and to use large-scale genomic and transcriptional analyses to study the molecular basis of acquired temozolomide resistance. Two independently obtained temozolomide-resistant variants exhibited no cross-resistance to other alkylating agents [1,3-bis(2-chloroethyl)-1-nitrosourea and carboplatin] and shared genetic alterations, such as loss of a 2p region and loss of amplification of chromosome 4 and 16q regions. The karyotypic alterations were compatible with clonal selection of preexistent resistant cells in the parental SNB-19 cell line. Microarray analysis showed that 78 out of 17,000 genes were differentially expressed between parental cells and both temozolomide-resistant variants. None are implicated in known resistance mechanisms, such as DNA repair, whereas interestingly, several genes involved in differentiation were down-regulated. The data suggest that the acquisition of resistance to temozolomide in this model resulted from the selection of less differentiated preexistent resistant cells in the parental tumor. [Mol Cancer Ther 2006;5(9):2182–92]

Gliomas are the most common brain tumor. They are usually treated by surgical resection combined with irradiation and alkylating agent–based chemotherapy. However, their prognosis remains poor. The therapeutic benefits obtained are transient and recurrence is the rule, together with resistance to chemotherapy. The limited efficacy of chemotherapy is generally attributed to two factors: intrinsic or acquired chemoresistance and the blood-brain barrier impeding the delivery of cytotoxic agents (1).

Temozolomide is a recently introduced, oral, and generally well-tolerated second-generation alkylating agent. Temozolomide has yielded a significant clinical benefit in high-grade gliomas (2). Temozolomide-induced O6-G-methylation is reversed by O6-methylguanine-DNA methyltransferase (MGMT) in a reaction leading to irreversible inactivation of the protein. Increased levels of MGMT or loss of the mismatch repair capacity confer resistance to temozolomide (3). However, some tumors display resistance to DNA-methylating drugs independently of either their MGMT level or their mismatch repair status, suggesting that other major mechanisms are involved in the resistance to alkylating agents, including the loss of a functional p53 which is able to induce cycle arrest and apoptosis (4), the p53 independent Chk1-mediated G2-M arrest which protects the cell from mitotic catastrophe (5), or the Akt pathway, frequently activated in glioblastoma, which bypass the temozolomide G2-M arrest on the one hand, and on the other hand, protects cells by suppressing senescence and mitotic catastrophe (6).

Comparative genomic hybridization (CGH) analysis of cell lines or primary tumors has revealed regions of chromosomal imbalances associated with acquired chemoresistance (79). None of these studies, however, concerned glioma cell lines and resistance to temozolomide.

In the present study, we used the temozolomide-sensitive SNB-19 cell line to generate temozolomide-resistant variants by culturing cells in the presence of incremental concentrations of temozolomide. The molecular alterations associated with acquired temozolomide resistance were identified using cytogenetic molecular techniques, including conventional cytogenetics, fluorescent in situ hybridization (FISH), CGH, and CGH arrays. Chromosomal regions having gained or lost DNA during the acquisition of resistance were disclosed. The additional use of microarray analysis allowed us to detect quantitative alterations of mRNA transcripts associated or not with genetic changes. We also evaluated the “kinetics” of these changes during the acquisition of temozolomide resistance.

Cell Culture, Treatment, and Isolation of Temozolomide-Resistant Variants

The glioma-derived cell line, SNB-19 (10), was cultivated in DMEM (Sigma, St. Quentin Fallavier, France) supplemented with 10% FCS (Dutscher, Brumath, France), penicillin G (102 IU/mL), streptomycin (50 μg/mL; Sigma), and l-glutamine (2 mmol/L; Sigma). Cells were maintained at 37°C in a humidified 5% CO2/95% air incubator, and harvested for passage with 0.5 mg/mL trypsin and EDTA (0.2 mg/mL) when they had reached confluence. For resistant cell selection, temozolomide (Interchim, Montluçon, France) was dissolved in DMSO as a 17 mmol/L stock solution, and diluted extemporaneously in complete medium at the desired concentration. At the first step of selection with 3 μmol/L of temozolomide, 12 clones were isolated from among >50, and were cultured separately or frozen. Two clones were treated with incremental concentrations of temozolomide (3, 5, 10, 20, 30, 60, and 150 μmol/L). At each step of selection, cells were exposed to a higher temozolomide concentration as soon as regrowth was apparent. Briefly, cells were plated in 25 cm2 flasks in their usual medium and allowed to attach overnight for drug selection, then the medium was replaced by 0.5 mL of temozolomide-containing fresh medium. After the first 2 hours, temozolomide-free fresh medium was added. 1,3-Bis(2-chloroethyl)-1-nitrosourea (carmustine, Bristol-Myers Squibb) was dissolved in ethanol at 17 mmol/L and carboplatin (Merck, Lyon, France) was diluted in water at 10 mg/mL.

Cell Proliferation Assay

Cell proliferation was determined by using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test (Sigma) and the methyl blue test. The amount of tetrazolium dye or methyl blue was proportional to the number of viable cells. Briefly, 5 × 103 cells were plated per well in 24-well polystyrene plates (ATGC, Noisy-le-Grand, France) in culture medium and allowed to adhere overnight. The medium was then replaced with fresh medium containing (or not) increasing concentrations of drug (0.03–150 μmol/L for the parental cells and 10–250 μmol/L for the variants) or the same concentration of the excipient (DMSO or ethanol) as that used for the highest drug dose used for 96 hours. For the MTT assay, the medium was replaced by MTT (2 mg/mL) diluted in medium (80%) for >3 hours and then replaced by DMSO, as previously described (11). For the methyl blue test assay, the surviving adherent cells were fixed with methanol and stained with methylene blue (1% in PBS buffer), and cell-fixed dye was eluted with 0.1 N of HCl (12). Absorbance (A) was measured in an automatic scanning photometer at a wavelength of 550 nm. Each experimental point was done in triplicate. The percentage of live cells was calculated as follows: P = (A in treated cells / A in control cells) × 100. Each assay was done at least thrice. Results are expressed as medians with the range. Statistical significance was determined using the Mann-Whitney U test (P ≤ 0.05).

Karyotype

Metaphases were harvested after a 2.5-hour colchicine block. Chromosome spreads were obtained according to previously described techniques (13). Karyotypes were established after R-banding.

DNA Extraction

DNA was extracted from frozen cells: samples were digested in extraction buffer (10 mmol/L Tris-Cl, 2 mmol/L EDTA, and 400 mmol/L NaCl) and 1.25% SDS and proteinase K (67 μg/mL) at 37°C overnight. DNA was subsequently treated with RNase (10 mg/mL) for 1 hour at 37°C. NaCl (5 mol/L) was added before precipitation with ethanol. The concentration and molecular size of DNA were estimated using a spectrophotometer (Uvikon 923; Fisher Bioblock Scientific, Illkirch, France) and ethidium bromide-stained agarose gels.

CGH and Digital Image Analysis

CGH experiments were done according to published protocols (14). DNA was labeled with fluorescein-12-dUTP and normal reference DNA obtained from normal lymphocytes, with Texas red-5-dUTP (Vysis, Rungis, France) using a commercially available nick-translation kit (Vysis). Four hundred nanograms of labeled tumor and reference DNA were coprecipitated with Cot-I DNA, and then denatured. Metaphase cell preparations obtained from normal lymphocytes were denatured and incubated with the probes in a humid chamber at 37°C for 3 days. Slides were washed and mounted in antifade solution. Digital images of the three fluorochromes were acquired using a computer-driven cooled CCD (Sensys, Photometrics, Evry, France) camera mounted on a fluorescence microscope (DMRB, Leica, Germany) equipped with selective single bandpass filters and interfaced to the CGH QUIPS software package (Vysis). Chromosomes stained with 4′,6-diamidino-2-phenylindole were identified by computer-generated reverse DAPI banding. The green to red fluorescence ratio along each chromosome was calculated by the appropriate software. CGH analysis of each tumor was done on 15 metaphases. The threshold values for losses and gains were set at 0.9 and 1.1, respectively. Amplification corresponded to a ratio exceeding 1.5.

CGH Arrays

A genome-wide resource of 3,342 FISH-mapped, sequenced BAC and PAC clones verified for gene and marker contents were represented as immobilized DNA targets on glass slides for array-based CGH analysis allowing a mean resolution of 1 Mb all along the genome. Each clone was spotted in triplicate on a slide with an Aminosilanne coating (Corning UltraGAPS, NH3+) with the Microgrid TAS BioRobotics spotter. These slides were a generous gift from the INSERM Unit U520 (France).

After extraction, 1.5 μg of each test and control DNA sample was digested with DpnII enzyme (Ozyme, Saint Quentin en Yvelines, France) and purified with a QIAquick PCR purification kit (Qiagen, Courtaboeuf, France). They were then labeled by random priming using a Bioprime DNA labeling kit (Invitrogen, Cergy Pontoise, France) with the appropriate cyanine dye (Cy3 or Cy5; Perkin-Elmer, Wellesley, MA). The control and test DNAs were coprecipitated with Cot-1 DNA (Invitrogen), denatured and resuspended in hybridization buffer (50% formamide). Competitive cohybridization was done on CGH array slides preblocked by succinic anhydride/N-methyl-2-pyrrolidinone/borate buffer (Sigma-Aldrich).

After a 24-hour hybridization, slides were washed with SDS and saline citrate, dried, and scanned using a 4000B scan (Axon Instruments, Union City, CA). Image analysis was done with Genepix 5.1 software (Axon Instruments) and processed using software developed at the Curie Institute.

Any BAC with less than two replicates flagged for not fulfilling qualitative spot criteria was excluded. A ratio of <0.8 was considered as a loss, a ratio of >1.2 as a gain, and a ratio of >1.5 as amplification.

Chromosome FISH

FISH was done as previously described (15). Several chromosome-specific biotinylated painting probes (Oncor Appligene, Illkirch, France) were hybridized to metaphase spreads. The biotinylated probes were detected by goat anti-biotin antibody (Vector Laboratories, Burlingame, CA) and fluoresceinated anti-goat rabbit antibody (Biosys, Compiègne, France). Chromosomes were counterstained with propidium iodide. Fluorescent signals were detected using an epifluorescence microscope (DMRB, Leica). Twenty metaphase spreads were analyzed to evaluate signal distribution.

RNA Extraction

Total RNA was isolated using an RNA extraction kit (Macherey-Nagel, Hoerdt, France) according to the manufacturer's instructions. RNA quality was assessed using an Agilent 2100 Bioanalyser (Massy, France).

Global Analysis of Gene Transcription

For probe preparation, 20 μg of RNA from sensitive or resistant cell lines were reverse-transcribed and labeled using the CyScribe Post-Labeling Kit procedure (Amersham Biosciences, Orsay, France), without modifications. After purification with the CyScribe GFX Purification Kit (Amersham Biosciences), the cDNAs were combined and applied to the arrays (Agilent human 1A oligonucleotide microarray), exactly as recommended by the manufacturer. Slides were scanned in both Cy3 and Cy5 channels with the GenePix 4000 scanner (Axon Instruments) and analyzed by GenePix Pro Software (Axon Instruments). For dye normalization, the LOWESS (locally weighted linear regression) method was applied, using the VARAN software.6

A log ratio of the red and green channel signals (local background subtracted) was obtained from the processed data. Dye-swap replicates were done for each experiment and spots were selected on the basis of a 2 SD cut/of the mean log ratios. Ratios of 2 and 0.5 were set as thresholds to identify differentially expressed genes. They were confirmed as being significantly underexpressed or overexpressed in resistant versus sensitive cells, as determined by significance analysis of microarrays, using a false discovery rate of <10%.

Analysis of Gene Expression

cDNA was prepared from each cell line using a combination of random primers (Promega, Charbonnières, France) and Superscript II (Invitrogen) for validation of array results by quantitative PCR. Oligos were designed around intron-exon boundaries for each gene using primer3 software.7

Each PCR was carried out in triplicate in a 20 μL volume using SybrGreen Mastermix (Applied Biosystems, Courtaboeuf, France) in the ABI Prism system. Each primer set was first tested to determine optimal concentrations, to assess the specificity of the PCR product, and to test PCR efficacy. Values for each gene were normalized to the expression levels of three housekeeping genes: HMBS, BM, and SDHA, and then a ratio comparing expression in resistant versus sensitive cell lines was calculated. The sequences of the primers used for real-time PCR are listed in Table 1.

Table 1.

List of genes verified by real-time PCR

Gene symbolRefSeq AccSequence of primersA4 R/SC1 R/SA4 QPCRC1 QPCR
BMPR1B NM_001203 TCAAGAAGTTACGCCCCTCAGTCAGCCTTGATGCAGGATT 0.39 0.38 0.13 0.01 
CTGF NM_001901 AGCCTCAATTTCTGAACACCACTCCCCTTTGCAAACAATCT 2.84 2.13 4.95 3.64 
CYP4F2 NM_001082 GCTTTGACCCAGAGAACATCAGCCAGGACCACCTTCATCT 2.54 2.39 Not determined 4.02 
EDNRB NM_000115 CCCTTTCCTTCTCCATGTCAAAAAGCCACTGAATGCAATTTT 0.35 0.37 0.32 0.26 
ELL NM_032245 ACCCCAGGTTTAAACGGAACTGTACTCGGCATTGAAGTCG 2.17 2.32 1.72 1.30 
EPHA3 NM_005233 TTCACGGGTGTGGAGTACAGACTGGGCCATTCTTTGATTG 0.41 0.19 0.70 0.12 
FOS NM_005252 AGAATCCGAAGGGAAAGGAACTTCTCCTTCAGCAGGTTGG 2.06 4.66 1.54 5.18 
GJA1 NM_000165 TCTTTTGGAGTGACCAGCAAAAGGCATTTGGAGAAACTGG 0.42 0.39 0.17 0.07 
GPM6A NM_005277 GGACCTTCGTCAGTTTGGAACTCCAGCAAGTGCCACAATA 0.44 0.39 0.37 0.16 
HTRA3 NM_053044 GACTTCCCAGAGGTCAGCAGCGTTGACCTTGACGATGATG 2.17 2.49 1.59 1.56 
IGFBP2 NM_000597 CCTCTACTCCCTGCACATCCGTTGGGGTTCACACACCAG 0.45 0.26 0.62 0.18 
IGFBP7 NM_001553 AGTGGTTGATGCCTTACATGACCATGACTACTTTTAACCATGCAG 0.39 0.14 0.58 0.07 
LEF1 NM_016269 CTTTATCCAGGCTGGTCTGCTCGTTTTCCACCATGTTTCA 0.42 0.41 0.60 0.34 
RAI NM_006663 GGGTGAAGCCTCAAAGGAGTTGCAGATAAAGGCAGCAAAA 2.35 2.45 6.90 5.08 
RGL NM_015149 TGGTCTTTCCAGGAGATTGGGCACAGAAGCACAAATCGAA 0.45 0.39 0.55 0.38 
SEC3L1 NM_018261 GCCAAACAAAAATACACAGATCACGAGCTTCAACACCTTCAAA 0.44 0.43 0.58 0.29 
SRP72 NM_006947 AGCAACTGCAGGAGCTTCATTGGTGTCTTGGGGGTATGAT 0.35 0.31 0.51 0.34 
SSB1 NM_025106 GCACAGGGTTGCATTTCTTTCTCGCCCTCCCTTCTTAGTT 2.48 2.05 3.35 3.96 
ZNF436 NM_030634 TGCACAGAGGGACCTTTACCTGCTTGGGATTTACCTCGTT 0.47 0.30 0.49 0.10 
Gene symbolRefSeq AccSequence of primersA4 R/SC1 R/SA4 QPCRC1 QPCR
BMPR1B NM_001203 TCAAGAAGTTACGCCCCTCAGTCAGCCTTGATGCAGGATT 0.39 0.38 0.13 0.01 
CTGF NM_001901 AGCCTCAATTTCTGAACACCACTCCCCTTTGCAAACAATCT 2.84 2.13 4.95 3.64 
CYP4F2 NM_001082 GCTTTGACCCAGAGAACATCAGCCAGGACCACCTTCATCT 2.54 2.39 Not determined 4.02 
EDNRB NM_000115 CCCTTTCCTTCTCCATGTCAAAAAGCCACTGAATGCAATTTT 0.35 0.37 0.32 0.26 
ELL NM_032245 ACCCCAGGTTTAAACGGAACTGTACTCGGCATTGAAGTCG 2.17 2.32 1.72 1.30 
EPHA3 NM_005233 TTCACGGGTGTGGAGTACAGACTGGGCCATTCTTTGATTG 0.41 0.19 0.70 0.12 
FOS NM_005252 AGAATCCGAAGGGAAAGGAACTTCTCCTTCAGCAGGTTGG 2.06 4.66 1.54 5.18 
GJA1 NM_000165 TCTTTTGGAGTGACCAGCAAAAGGCATTTGGAGAAACTGG 0.42 0.39 0.17 0.07 
GPM6A NM_005277 GGACCTTCGTCAGTTTGGAACTCCAGCAAGTGCCACAATA 0.44 0.39 0.37 0.16 
HTRA3 NM_053044 GACTTCCCAGAGGTCAGCAGCGTTGACCTTGACGATGATG 2.17 2.49 1.59 1.56 
IGFBP2 NM_000597 CCTCTACTCCCTGCACATCCGTTGGGGTTCACACACCAG 0.45 0.26 0.62 0.18 
IGFBP7 NM_001553 AGTGGTTGATGCCTTACATGACCATGACTACTTTTAACCATGCAG 0.39 0.14 0.58 0.07 
LEF1 NM_016269 CTTTATCCAGGCTGGTCTGCTCGTTTTCCACCATGTTTCA 0.42 0.41 0.60 0.34 
RAI NM_006663 GGGTGAAGCCTCAAAGGAGTTGCAGATAAAGGCAGCAAAA 2.35 2.45 6.90 5.08 
RGL NM_015149 TGGTCTTTCCAGGAGATTGGGCACAGAAGCACAAATCGAA 0.45 0.39 0.55 0.38 
SEC3L1 NM_018261 GCCAAACAAAAATACACAGATCACGAGCTTCAACACCTTCAAA 0.44 0.43 0.58 0.29 
SRP72 NM_006947 AGCAACTGCAGGAGCTTCATTGGTGTCTTGGGGGTATGAT 0.35 0.31 0.51 0.34 
SSB1 NM_025106 GCACAGGGTTGCATTTCTTTCTCGCCCTCCCTTCTTAGTT 2.48 2.05 3.35 3.96 
ZNF436 NM_030634 TGCACAGAGGGACCTTTACCTGCTTGGGATTTACCTCGTT 0.47 0.30 0.49 0.10 

NOTE: Results are expressed as the resistant/sensitive ratios of expression. The first two columns (A4 R/S and C1 R/S) are the results of microarray experiments for SNB-19-A4 and SNB-19-C1, respectively. The last two columns (A4 QPCR and C1 QPCR) show corresponding real-time quantitative PCR results.

In addition to these genes, MGMT expression was also tested at the RNA level using the following primers: 5′-CCTGGCTGAATGCCTATTTC-3′ (forward) and 5′-GATGAGGATGGGGACAGGATT-3′ (reverse). The methylation status of the MGMT promoter was also evaluated, as previously described (16).

Acquisition of Resistance to Temozolomide by the SNB-19 Glioma Cell Line

SNB-19 parental cells were very sensitive to temozolomide (Table 2). Two clones of resistant variants (SNB-19A4 and SNB-19C1) were isolated at an early stage of drug treatment (3 μmol/L of temozolomide) and then successively exposed to incremental doses of temozolomide (up to 150 μmol/L). After initial killing of a large majority of cells, surviving cells displayed a normal rate of proliferation. They were then submitted to a new selection step up to 150 μmol/L. The IC50 of temozolomide was evaluated by both MTT and methyl blue test techniques, and similar results were obtained. The SNB-19A4 and SNB-19C1 variants showed a 100-fold (P = 0.02) and 55-fold (P = 0.008) stronger resistance to temozolomide than the parental SNB-19 cell line, respectively. After 20 passages in temozolomide-free medium, the temozolomide resistance factor remained constant.

Table 2.

Comparative cytotoxicity of temozolomide, 1,3-bis(2-chloroethyl)-1-nitrosourea, and carboplatin in SNB-19 and its temozolomide-resistant sublines, SNB-19A4 and SNB-19C1

DrugsCells, IC50 median (range)
SNB-19SNB-19A4*PSNB-19C1*P
Temozolomide (μmol/L) 1.03 (0.042–1.7) 101 (90–113) 0.02 55 (40–59) 0.008 
1,3-Bis(2-chloroethyl)-1-nitrosourea (μmol/L) 9.5 (8–13) 21 (13–29) 0.038 16 (13–19) 0.037 
Carboplatin (μg/L) 3.1 (0.58–5.8) 13 (5.5–15) 0.12 3.1 (1.8–6) 0.65 
DrugsCells, IC50 median (range)
SNB-19SNB-19A4*PSNB-19C1*P
Temozolomide (μmol/L) 1.03 (0.042–1.7) 101 (90–113) 0.02 55 (40–59) 0.008 
1,3-Bis(2-chloroethyl)-1-nitrosourea (μmol/L) 9.5 (8–13) 21 (13–29) 0.038 16 (13–19) 0.037 
Carboplatin (μg/L) 3.1 (0.58–5.8) 13 (5.5–15) 0.12 3.1 (1.8–6) 0.65 
*

Selected at 150 μmol/L of temozolomide.

Statistical significance was determined by the Mann-Whitney test.

Nonsignificant.

The resistance of both variants to different alkylating agents was compared with that of the parental cells. The IC50 of 1,3-bis(2-chloroethyl)-1-nitrosourea was increased 2.2-fold (P = 0.038) and 1.6-fold (P = 0.037) for SNB-19A4 and SNB-19C1, respectively. The IC50 of carboplatin was increased 4.1-fold (P = 0.12) for SNB-19A4 and remained stable at 3.1 μmol/L for SNB-19C1 (Table 2). Thus, the resistance to temozolomide acquired by these two variants was stable and could be considered specific to temozolomide.

Genetic Alterations of Temozolomide-Resistant Cell Lines

CGH was used to analyze the alterations in SNB-19A4 and SNB-19C1 variants. SNB-19 showed numerous rearrangements (Fig. 1A), as previously published (10). DNA was extracted from resistant cells at the highest temozolomide concentration (150 μmol/L) and hybridized with normal DNA (data not shown). Most of the numerous imbalances observed were shared by the parental cells and the temozolomide-resistant cells lines. To eliminate these shared imbalances, the DNA from each resistant variant was cohybridized with the DNA from the parental SNB-19 cells. We were therefore able to identify alterations in the temozolomide-resistant clones that were absent in the parental cell line. Such alterations concerned the loss of a 2p region involving the 2p16-2p25 bands, loss of partial amplification of the 4p14-4q21 region and loss of amplification of the 16q12-16q22 region (Fig. 1B and C); these last two regions appeared with a normal ratio during hybridization versus control DNA. During a second step, the same cohybridizations were done with DNA extracted at a lower level of temozolomide resistance (3, 30, and 60 μmol/L). These alterations were present as early as the 30 μmol/L dose of temozolomide but were not detected at the 3 μmol/L dose (data not shown). The IC50 of temozolomide for the SNB-19C1 variant selected at 30 μmol/L of temozolomide was the same as that for the variant selected at 150 μmol/L (IC50 = 45 μmol/L).

Figure 1.

CGH profiles of SNB-19 cells and of SNB-19A4 and SNB-19C1 temozolomide-resistant variants. SNB-19 DNA versus normal control DNA (A), SNB-19A4 versus SNB-19 (B), and SNB-19C1 versus SNB-19 (C). Chromosome ideogram shows a ratio of 1 (central line), a ratio threshold of 0.9 for underrepresentation (left line), and 1.1 for overrepresentation (right line) of DNA/chromosome content. A, SNB-19 cells exhibit many chromosomal imbalances including amplification in chromosomes 4 and 16. Many of these rearrangements are frequently described in gliomas: e.g., gain of the PDGFRA region (on chromosome 4), gain of 7, loss of 10p, 10q and 13. B and C, in both temozolomide-resistant variants, a 2p region and the amplifications present on chromosomes 4 and 16 are lost.

Figure 1.

CGH profiles of SNB-19 cells and of SNB-19A4 and SNB-19C1 temozolomide-resistant variants. SNB-19 DNA versus normal control DNA (A), SNB-19A4 versus SNB-19 (B), and SNB-19C1 versus SNB-19 (C). Chromosome ideogram shows a ratio of 1 (central line), a ratio threshold of 0.9 for underrepresentation (left line), and 1.1 for overrepresentation (right line) of DNA/chromosome content. A, SNB-19 cells exhibit many chromosomal imbalances including amplification in chromosomes 4 and 16. Many of these rearrangements are frequently described in gliomas: e.g., gain of the PDGFRA region (on chromosome 4), gain of 7, loss of 10p, 10q and 13. B and C, in both temozolomide-resistant variants, a 2p region and the amplifications present on chromosomes 4 and 16 are lost.

Close modal

CGH array analyses were done with the same cell lines, first cohybridized with normal cell DNA (Fig. 2A), then again cohybridized between the DNA of each resistant variant (DNA extracted at 150 μmol/L temozolomide) and the DNA of the parental SNB-19 cells (Fig. 2B and C). These results confirmed those of the conventional metaphase-based CGH analysis. However, the altered regions were more accurately defined (2p16.1-2p25.3, 4p14.4-4q21.22, and 16q12.1-16q22.1) and further loss of amplification of the 1p13.2-1q21.1 region shared by SNB-19A4 and SNB-19C1 was detected.

Figure 2.

CGH array profiles of SNB-19 cells versus a normal control (A) and of temozolomide-resistant variants SNB-19A4 (B) and SNB-19C1 (C) both versus SNB-19. Each point corresponds to a BAC on the slide. They are colored according to the ratio of fluorescence. When the ratio was <0.8 (corresponding to a loss), the point is in green, and in red when the ratio was >1.2 (gain or amplification). Between these two ratios, the points are in yellow. As also shown by CGH, SNB-19 exhibited numerous imbalances, whereas some additional ones were not detected by CGH. Among them, gain of 1q32 is also frequent. Loss of 2p, 4, and 16q are also shown by CGH array profiles.

Figure 2.

CGH array profiles of SNB-19 cells versus a normal control (A) and of temozolomide-resistant variants SNB-19A4 (B) and SNB-19C1 (C) both versus SNB-19. Each point corresponds to a BAC on the slide. They are colored according to the ratio of fluorescence. When the ratio was <0.8 (corresponding to a loss), the point is in green, and in red when the ratio was >1.2 (gain or amplification). Between these two ratios, the points are in yellow. As also shown by CGH, SNB-19 exhibited numerous imbalances, whereas some additional ones were not detected by CGH. Among them, gain of 1q32 is also frequent. Loss of 2p, 4, and 16q are also shown by CGH array profiles.

Close modal

Cytogenetic Alterations of Temozolomide-Resistant Variants as Studied by FISH and Karyotype Analysis

SNB-19 exhibited a hyperdiploid karyotype with 59 to 61 chromosomes. SNB-19A4 was also hyperdiploid with 54 to 55 chromosomes and SNB-19C1 was hypopentaploid with 102 to 107 chromosomes (data not shown).

FISH was done with chromosomal painting probes for chromosomes 2, 4, and 16 on metaphases because CGH showed shared rearrangements involving these three chromosomes. FISH revealed a derivative chromosome in the parental SNB-19 cell line, that involved chromosomes 2, 4, and 16 with at least five breakpoints (Fig. 3). This derivative chromosome was lost in both SNB-19A4-resistant and SNB-19C1-resistant variants. However, it was difficult to assign the lost chromosomal material found by CGH to the loss of this marker alone because several chromosomes were hybridized to these probes.

Figure 3.

Gray scale inverted DAPI banding (A) and FISH analysis of metaphase chromosomes of SNB-19 parental tumors. Chromosomes were counterstained with propidium iodide (red). FISH analysis of metaphases of the parental cell line with human chromosome painting of chromosomes 2 (B), 4 (C), and 16 (D) show that the submetacentric marker chromosome is a complex derivative chromosome comprising regions of chromosomes 2, 4, 16 and other unidentified chromosomes.

Figure 3.

Gray scale inverted DAPI banding (A) and FISH analysis of metaphase chromosomes of SNB-19 parental tumors. Chromosomes were counterstained with propidium iodide (red). FISH analysis of metaphases of the parental cell line with human chromosome painting of chromosomes 2 (B), 4 (C), and 16 (D) show that the submetacentric marker chromosome is a complex derivative chromosome comprising regions of chromosomes 2, 4, 16 and other unidentified chromosomes.

Close modal

Gene Expression Alterations in Temozolomide-Resistant Variants as Studied by Microarray Analysis

We did a comparative transcriptome analysis between parental SNB-19 cells and the two temozolomide-resistant variants. RNA isolated from SNB-19A4 and SNB-19C1 was cohybridized with RNA isolated from SNB-19 parental cells in a dye-swap experiment using Agilent arrays. The search for differential expression evidenced quantitative alterations of 245 genes between SNB-19A4 and SNB-19, and 284 genes between SNB-19C1 and SNB-19. By cross-checking the two lists of genes, 78 genes were found to exhibit similar alterations in the two temozolomide-resistant variants. Differential expression was confirmed by quantitative PCR analysis for 18 out of these 78 genes (Table 2).

Quantitative alterations of gene expression and genomic alterations were confronted. The location of the gene was known in 72 out of the 78 differentially expressed genes (Table 3; Fig. 4). Among them, 11 genes (15%) were located in the rearranged region (Table 3).

Table 3.

Differentially expressed genes in SNB-19A4 and SNB-19C1 versus SNB-19 ranging according to level of expression, physical mapping, and involvement in differentiation or stem cell apoptosis (A), mismatch repair or drug resistance (B), or oncogenesis or tumor progression (C)

Gene symbolNameSNB-19A4*SNB-19C1*Physical mappingFunction
CNN1 Calponin 1, basic, smooth muscle 3.2 2.27 19 AC 
KIAA1189 KIAA1189 protein 3.07 2.04  
CTGF Connective tissue growth factor 2.84 2.13  
GNA14 Guanine nucleotide binding protein (G protein), α 14 2.8 2.08  
PF4 Platelet factor 4 2.68 2.1  
PTGIR Prostaglandin I2 (prostacyclin) receptor (IP) 2.65 2.16 19  
OR4K11P OR4K11P 2.64 2.45 21  
CYP4F2 CYP4F2 2.54 2.39 19 
SSB1 SPRY domain-containing SOCS box protein SSB-1 2.48 2.05  
PLAC8 Placenta-specific 8 2.47 2.51 12 
ZPBP Zona pellucida binding protein 2.47 2.1  
KRT14 Keratin 14 (epidermolysis bullosa simplex, Dowling-Meara, Koebner) 2.46 2.53 17  
OR6T1 OR6T1 2.44 2.02 11  
FLJ44682 FLJ44682 2.39 2.31   
RAI RelA-associated inhibitor 2.35 2.45 19 
PRODH Proline dehydrogenase (oxidase) 1 2.33 2.5 22 
CST9L Cystatin 9-like (mouse) 2.24 2.02 20  
FZD2 Frizzled homologue 2 (Drosophila) 2.22 2.32 17 
SLC22A1 Solute carrier family 22 (organic cation transporter), member 1 2.19 2.26  
CEBPB CCAAT/enhancer binding protein (C/EBP), β 2.19 3.12 20 AC 
ELL Elongation factor RNA polymerase II 2.17 2.32 19 
HTRA3 Serine protease HTRA3 2.17 2.49  
IAPP Islet amyloid polypeptide 2.16 2.1 12  
TAS1R3 TAS1R3 2.13 2.23  
LIF Leukemia inhibitory factor (cholinergic differentiation factor) 2.09 2.53 22 
FOS v-fos FBJ murine osteosarcoma viral oncogene homologue 2.06 4.66 14 
PTPRCAP  2.01 2.35 11  
FLJ13852 Hypothetical protein FLJ13852 2.01 2.02  
C40 Hypothetical protein C40 0.5 0.45  
NEDD9 Neural precursor cell expressed, developmentally down-regulated 9 0.5 0.3 
TGFB3 Transforming growth factor, β3 0.49 0.49 14 
TPD52L1 Tumor protein D52-like 1 0.48 0.36  
NET-7 Transmembrane 4 superfamily member tetraspan NET-7 0.48 0.47 10  
HOMER1 Homer homologue 1 (Drosophila) 0.48 0.38  
ENSA Endosulfine α 0.48 0.5  
JDP1 J domain containing protein 1 0.47 0.31 10  
ZNF436 Zinc finger protein 436 0.47 0.3  
GALNT5 UDP-N-acetyl-α-d-galactosamine:polypeptide N-acetylgalactosaminyltransferase 5 (GalNAc-T5) 0.46 0.44  
PHKB Phosphorylase kinase, β 0.45 0.39 16  
BCMP11 Breast cancer membrane protein 11 0.45 0.43 
HADHA Hydroxyacyl-CoA dehydrogenase/α subunit 0.45 0.39  
IGFBP2 Insulin-like growth factor binding protein 2, 36 kDa 0.45 0.26 
CTSL2 Cathepsin L2 0.45 0.36 
RGL RalGDS-like gene 0.45 0.39  
ATP1A3 ATPase 0.45 0.4 19  
KLHL13 Kelch-like 13 (Drosophila) 0.45 0.37  
GPM6A Glycoprotein M6A 0.44 0.39 
CTNND2 Catenin (cadherin-associated protein), δ2 (neural plakophilin-related arm-repeat protein) 0.44 0.37  
KIAA1212 KIAA1212 0.44 0.41  
SEC3L1 SEC3-like 1 (S. cerevisiae0.44 0.43 
FEZ1 Fasciculation and elongation protein ζ1 (zygin I) 0.44 0.33 11 AC 
SLITRK6 SLIT and NTRK-like family, member 6 0.44 0.36 13 
NMU Neuromedin U 0.43 0.38  
GPR48 G protein-coupled receptor 48 0.43 0.29 11  
CLK1 CDC-like kinase 1 0.43 0.37 
PRPF4B PRP4 pre-mRNA processing factor 4 homologue B (yeast) 0.42 0.44  
GJA1 Gap junction protein, α1, 43 kDa (connexin 43) 0.42 0.39 ABC 
AFP α-Fetoprotein 0.42 0.36 
LEF1 Lymphoid enhancer-binding factor 1 0.42 0.41 
EPHA3 EphA3 0.41 0.19 
FBLN5 Fibulin 5 0.4 0.33 14 
TRIM22 Tripartite motif-containing 22 0.4 0.34 11 
IGFBP7 Insulin-like growth factor binding protein 7 0.39 0.14 
G3BP2 Ras-GTPase activating protein SH3 domain-binding protein 2 0.39 0.35  
HELLS Helicase, lymphoid-specific 0.39 0.48 10 BC 
BMPR1B Bone morphogenetic protein receptor, type IB 0.39 0.38 
SCNN1A Sodium channel, non–voltage-gated 1α 0.38 0.47 12  
GRIK2 Glutamate receptor, ionotropic, kainate 2 0.38 0.39 
C9orf58 Chromosome 9 open reading frame 58 0.38 0.41  
KIAA1764 KIAA1764 protein 0.38 0.31  
TKTL1 Transketolase-like 1 0.37 0.45  
BICD1 Bicaudal D homologue 1 (Drosophila) 0.37 0.43 12  
SRP72 Signal recognition particle, 72 kDa 0.35 0.31 
EDNRB Endothelin receptor type B 0.35 0.37 13 
CAPS Calcyphosine 0.33 0.33 19 
LOC83690 CocoaCrisp 0.32 0.23  
PLEKHB1 Pleckstrin homology domain containing, family B (evectins) member 1 0.3 0.27 11 AC 
ARHGEF4 Rho guanine nucleotide exchange factor (GEF) 4 0.28 0.3  
Gene symbolNameSNB-19A4*SNB-19C1*Physical mappingFunction
CNN1 Calponin 1, basic, smooth muscle 3.2 2.27 19 AC 
KIAA1189 KIAA1189 protein 3.07 2.04  
CTGF Connective tissue growth factor 2.84 2.13  
GNA14 Guanine nucleotide binding protein (G protein), α 14 2.8 2.08  
PF4 Platelet factor 4 2.68 2.1  
PTGIR Prostaglandin I2 (prostacyclin) receptor (IP) 2.65 2.16 19  
OR4K11P OR4K11P 2.64 2.45 21  
CYP4F2 CYP4F2 2.54 2.39 19 
SSB1 SPRY domain-containing SOCS box protein SSB-1 2.48 2.05  
PLAC8 Placenta-specific 8 2.47 2.51 12 
ZPBP Zona pellucida binding protein 2.47 2.1  
KRT14 Keratin 14 (epidermolysis bullosa simplex, Dowling-Meara, Koebner) 2.46 2.53 17  
OR6T1 OR6T1 2.44 2.02 11  
FLJ44682 FLJ44682 2.39 2.31   
RAI RelA-associated inhibitor 2.35 2.45 19 
PRODH Proline dehydrogenase (oxidase) 1 2.33 2.5 22 
CST9L Cystatin 9-like (mouse) 2.24 2.02 20  
FZD2 Frizzled homologue 2 (Drosophila) 2.22 2.32 17 
SLC22A1 Solute carrier family 22 (organic cation transporter), member 1 2.19 2.26  
CEBPB CCAAT/enhancer binding protein (C/EBP), β 2.19 3.12 20 AC 
ELL Elongation factor RNA polymerase II 2.17 2.32 19 
HTRA3 Serine protease HTRA3 2.17 2.49  
IAPP Islet amyloid polypeptide 2.16 2.1 12  
TAS1R3 TAS1R3 2.13 2.23  
LIF Leukemia inhibitory factor (cholinergic differentiation factor) 2.09 2.53 22 
FOS v-fos FBJ murine osteosarcoma viral oncogene homologue 2.06 4.66 14 
PTPRCAP  2.01 2.35 11  
FLJ13852 Hypothetical protein FLJ13852 2.01 2.02  
C40 Hypothetical protein C40 0.5 0.45  
NEDD9 Neural precursor cell expressed, developmentally down-regulated 9 0.5 0.3 
TGFB3 Transforming growth factor, β3 0.49 0.49 14 
TPD52L1 Tumor protein D52-like 1 0.48 0.36  
NET-7 Transmembrane 4 superfamily member tetraspan NET-7 0.48 0.47 10  
HOMER1 Homer homologue 1 (Drosophila) 0.48 0.38  
ENSA Endosulfine α 0.48 0.5  
JDP1 J domain containing protein 1 0.47 0.31 10  
ZNF436 Zinc finger protein 436 0.47 0.3  
GALNT5 UDP-N-acetyl-α-d-galactosamine:polypeptide N-acetylgalactosaminyltransferase 5 (GalNAc-T5) 0.46 0.44  
PHKB Phosphorylase kinase, β 0.45 0.39 16  
BCMP11 Breast cancer membrane protein 11 0.45 0.43 
HADHA Hydroxyacyl-CoA dehydrogenase/α subunit 0.45 0.39  
IGFBP2 Insulin-like growth factor binding protein 2, 36 kDa 0.45 0.26 
CTSL2 Cathepsin L2 0.45 0.36 
RGL RalGDS-like gene 0.45 0.39  
ATP1A3 ATPase 0.45 0.4 19  
KLHL13 Kelch-like 13 (Drosophila) 0.45 0.37  
GPM6A Glycoprotein M6A 0.44 0.39 
CTNND2 Catenin (cadherin-associated protein), δ2 (neural plakophilin-related arm-repeat protein) 0.44 0.37  
KIAA1212 KIAA1212 0.44 0.41  
SEC3L1 SEC3-like 1 (S. cerevisiae0.44 0.43 
FEZ1 Fasciculation and elongation protein ζ1 (zygin I) 0.44 0.33 11 AC 
SLITRK6 SLIT and NTRK-like family, member 6 0.44 0.36 13 
NMU Neuromedin U 0.43 0.38  
GPR48 G protein-coupled receptor 48 0.43 0.29 11  
CLK1 CDC-like kinase 1 0.43 0.37 
PRPF4B PRP4 pre-mRNA processing factor 4 homologue B (yeast) 0.42 0.44  
GJA1 Gap junction protein, α1, 43 kDa (connexin 43) 0.42 0.39 ABC 
AFP α-Fetoprotein 0.42 0.36 
LEF1 Lymphoid enhancer-binding factor 1 0.42 0.41 
EPHA3 EphA3 0.41 0.19 
FBLN5 Fibulin 5 0.4 0.33 14 
TRIM22 Tripartite motif-containing 22 0.4 0.34 11 
IGFBP7 Insulin-like growth factor binding protein 7 0.39 0.14 
G3BP2 Ras-GTPase activating protein SH3 domain-binding protein 2 0.39 0.35  
HELLS Helicase, lymphoid-specific 0.39 0.48 10 BC 
BMPR1B Bone morphogenetic protein receptor, type IB 0.39 0.38 
SCNN1A Sodium channel, non–voltage-gated 1α 0.38 0.47 12  
GRIK2 Glutamate receptor, ionotropic, kainate 2 0.38 0.39 
C9orf58 Chromosome 9 open reading frame 58 0.38 0.41  
KIAA1764 KIAA1764 protein 0.38 0.31  
TKTL1 Transketolase-like 1 0.37 0.45  
BICD1 Bicaudal D homologue 1 (Drosophila) 0.37 0.43 12  
SRP72 Signal recognition particle, 72 kDa 0.35 0.31 
EDNRB Endothelin receptor type B 0.35 0.37 13 
CAPS Calcyphosine 0.33 0.33 19 
LOC83690 CocoaCrisp 0.32 0.23  
PLEKHB1 Pleckstrin homology domain containing, family B (evectins) member 1 0.3 0.27 11 AC 
ARHGEF4 Rho guanine nucleotide exchange factor (GEF) 4 0.28 0.3  

NOTE: Genes exhibiting a change in copy number and in expression level are in boldface.

*

Ratio of expression in the resistant variant versus SNB-19.

Figure 4.

Chromosomal location of the expression of modified genes. Most of them are located on chromosomes 2, 4, and 16, chromosomes involved in the rearrangement variants had in common, as well as on chromosomes 1, 6, 11, and mainly 19. Most of them were underexpressed.

Figure 4.

Chromosomal location of the expression of modified genes. Most of them are located on chromosomes 2, 4, and 16, chromosomes involved in the rearrangement variants had in common, as well as on chromosomes 1, 6, 11, and mainly 19. Most of them were underexpressed.

Close modal

MGMT Expression

In order to test whether MGMT expression might be responsible for part of the resistance mechanism in A4 and C1 variants, we tested MGMT expression at the RNA level, but it was not detectable. Analysis of the methylation status of the MGMT promoter showed that it was fully methylated in all cell lines. Consequently, the MGMT level did not allow us to discriminate SNB-19 from the resistant variants.

Malignant glial tumors are well known to be refractory to chemotherapy. Significant clinical benefits have been obtained with temozolomide, a recently introduced alkylating agent (2). The main purpose of the present study was to generate temozolomide-resistant variants from SNB-19, a malignant glioma cell line, in an attempt to identify the genetic alterations associated with acquired resistance to temozolomide. Several temozolomide-resistant variants were obtained by stepwise selection with incremental doses of temozolomide in the culture medium. Genomic alterations appeared as early as the 30 μmol/L concentration, which roughly corresponds with the mean peak concentration obtained after conventional treatment (i.e., 150 mg/m2; ref. 17).

The resistance of these two variants can be considered specific to temozolomide given the difference in the P value between temozolomide and 1,3-bis(2-chloroethyl)-1-nitrosourea and no cross-resistance was noted with carboplatin, a third alkylating agent also used to treat glioblastoma-bearing patients. This was surprising, because resistance to a given alkylating agent often extends to other alkylating agents and even to other drugs (18, 19). Ma et al. (18) found that tumor cell–acquired resistance to temozolomide was associated with increased MGMT activity and alterations of apoptosis-controlling gene, i.e., increased Bcl2 and BclXL and decreased Bad, Bax, and BclXs. However, none of these genes were identified in our microarray analysis.

Hypermethylation of the MGMT CpG island can cause transcriptional silencing in cell lines unable to repair O6-methyl (20). We found no changes on MGMT expression which stay undetectable. The promoter methylation was also unchanged, with both parental cell lines and resistant variants giving 100% methylation (data not shown). A mismatch repair deficiency has not been addressed specifically but such an abnormality is infrequent in gliomas. A differential expression was not found on MGMT genes, mismatch repair genes, or other genes involved in drug resistance mechanisms including base excision repair (21), drug-metabolism, and detoxification (mainly through higher levels of glutathione or glutathione-S-transferase; ref. 22).

Resistance to temozolomide also involves p53 status, Chk1- and Chk2-mediated G2 checkpoint pathways, and Akt activation. In response to temozolomide, p53 induces cell cycle arrest and apoptosis (4). On the other hand, MGMT inactivation may facilitate AT-GC mutations on p53 (23, 24), explaining the correlation between MGMT inactivation and p53 dysfunction (25, 26). In response to temozolomide, Chk1 induces G2 arrest preventing from cell death independently of p53 status (5). Akt pathway activation, which is a frequent feature in glioblastoma, bypasses these mechanisms, preventing both G2 arrest and cell death (6). Again, none of the genes differentially expressed in our microarray analysis involved these pathways.

Gliomas are known to display very rearranged karyotypes with numerous chromosomal imbalances, due to sequential chromosomal endoreduplications and losses of chromosomal material (13). Among the imbalances found in SNB-19, some are frequent in gliomas: gain of 1q32 and gain of the chromosome 4 region bearing the PDGFRA gene (15, 27).

SNB-19 and the two resistant variants were aneuploid and their karyotypes were very complex, but strikingly, there were fewer rearrangements in the resistant variants. The two temozolomide-resistant variants harbored genomic differences with the parental SNB-19 cell line: loss of a 2p region and loss of amplification of the chromosome 4 and 16q regions. This observation does not fit with the commonly accepted hypothesis assuming that the acquisition of drug resistance, evolving through accidental chromosome recombinations, harbors more chromosomal rearrangements (28).

A likely explanation is that temozolomide-resistant variants with less rearranged genomes preexisted in the parental cell line and were selected through exposure to temozolomide. This could be related to cell passaging, but these clones were cultured totally independently after the first 3 μmol/L step. Moreover, two additional variants displaying the same alterations were further isolated (data not shown). The less complex rearrangement pattern of the temozolomide-resistant variants indicates that they may have more in common with an ancestral genotype than the temozolomide-sensitive parental cells, which is compatible with the idea that chemoresistance resulted from the enrichment of preexistent cells.

The expression of 17,000 genes was quantified by microarray analysis. Seventy-eight genes seemed to be differentially expressed in both variants: 15% were located in regions that had sustained losses (11). Among the 78 genes, only two, CLK1 and RAI, were previously associated with a response to cisplatin treatment (29, 30). However, none were associated with resistance to temozolomide. Thirty (19 underexpressed and 11 overexpressed) of these 78 genes seemed to be of specific interest because of their function.

Among the underexpressed genes, some have been described as tumor suppressor genes in solid tumor, IGFBP7 (31), FEZ1/LZTS1 (32), and GJA1 (33), or as inductors of apoptosis, such as CTGF (34). Many are involved in differentiation, notably in the nervous system, or in embryonic pathways such as GJA1 (35), GPM6A (36), FBLN-5 (37), NEDD9 (38), BMPR1B (39), AFP (40), EPHA3 (41), CYP4F2 (42), SEC3L1 (43), HELLS (44, 45), EDNRB (46), and PLEKHB1 (47). PLEKHB1 has recently been found to be down-regulated in grade 4 (glioblastomas) versus grade 1 gliomas (pilocytic astrocytoma). LEF1 is a transcription factor inhibiting the Wnt pathway, a key regulator of cell development and of the stem cell pool (48).

Several overexpressed genes in our temozolomide-resistant variants are involved in developmental pathways such as FZD2 (implicated in the Wnt pathway; ref. 48), or in stem cell self-renewal and proliferation such as LIF (49, 50), ELL (51), and CNN1 (52).

Thus, a group of differentially expressed genes in the SNB-19 temozolomide-resistant variants points to a shift towards a cancer stem cell–like genotype. The concept of a cancer stem cell was recently explored in human brain tumors and cell lines (53, 54). Tumor stem cells retain the ability to self-renew and might explain why many cancers relapse under conditions in which most tumor cells seem to be killed following therapy (54). An emerging model proposes that tumor stem cells are naturally resistant to chemotherapy through their quiescence, their capacity for DNA repair and increased ABC transporter expression (55). In conclusion, our cytogenetic analyses suggest that acquired temozolomide resistance in this malignant glioma model involves the selection of preexistent resistant cell variants. The gene expression profile of temozolomide-resistant variants displayed a complex picture with decreased expression of genes involved in differentiation and increased expression of genes known to be expressed in stem cells. Altogether, these data suggest that acquired temozolomide resistance could have resulted from the selection of less differentiated preexistent resistant cells with stem cell–like characteristics in the parental tumor.

Grant support: Ligue National contre le Cancer d'Ille et Vilaine.

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.

We thank Olivier Delattre for his generous gift of the CGH array slides and for granting access to his analysis platform; Dr. Jean-Gabriel Judde for critical reading of the manuscript; Dr. François Apiou, Anne-Marie-Dutrillaux, and Michèle Gerbeault for their technical assistance in karyotype, FISH, and CGH analyses; and Lorna Saint Ange (Institut Gustave-Roussy) for editing.

1
Fortin D. The blood-brain barrier should not be underestimated in neuro-oncology.
Rev Neurol (Paris)
2004
;
160
:
523
–32.
2
Stupp R, van den Bent MJ, Hegi ME. Optimal role of temozolomide in the treatment of malignant gliomas.
Curr Neurol Neurosci Rep
2005
;
5
:
198
–206.
3
Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia.
Cancer Res
1999
;
59
:
793
–7.
4
Bocangel DB, Finkelstein S, Schold SC, Bhakat KK, Mitra S, Kokkinakis DM. Multifaceted resistance of gliom as to temozolomide.
Clin Cancer Res
2002
;
8
:
2725
–34.
5
Hirose Y, Berger MS, Pieper RO. Abrogation of the Chk1-mediated G(2) checkpoint pathway potentiates temozolomide-induced toxicity in a p53-independent manner in human glioblastoma cells.
Cancer Res
2001
;
61
:
5843
–9.
6
Hirose Y, Katayama M, Mirzoeva OK, Berger MS, Pieper RO. Akt activation suppresses Chk2-mediated, methylating agent-induced G2 arrest and protects from temozolomide-induced mitotic catastrophe and cellular senescence.
Cancer Res
2005
;
65
:
4861
–9.
7
Wasenius VM, Jekunen A, Monni O, et al. Comparative genomic hybridization analysis of chromosomal changes occurring during development of acquired resistance to cisplatin in human ovarian carcinoma cells.
Genes Chromosomes Cancer
1997
;
18
:
286
–91.
8
Rao PH, Houldsworth J, Palanisamy N, et al. Chromosomal amplification is associated with cisplatin resistance of human male germ cell tumors.
Cancer Res
1998
;
58
:
4260
–3.
9
Yasui K, Mihara S, Zhao C, et al. Alteration in copy numbers of genes as a mechanism for acquired drug resistance.
Cancer Res
2004
;
64
:
1403
–10.
10
Gross JL, Behrens DL, Mullins DE, Kornblith PL, Dexter DL. Plasminogen activator and inhibitor activity in human glioma cells and modulation by sodium butyrate.
Cancer Res
1998
;
48
:
291
–6.
11
Dimanche-Boitrel MT, Pelletier H, Genne P, et al. Confluence-dependent resistance in human colon cancer cells: role of reduced drug accumulation and low intrinsic chemosensitivity of resting cells.
Int J Cancer
1992
;
50
:
677
–82.
12
Raynaud S, Nemati F, Miccoli L, et al. Antitumoral effects of squamocin on parental and multidrug resistant MCF7 (human breast adenocarcinoma) cell lines.
Life Sci
1999
;
65
:
525
–33.
13
Pruchon E, Chauveinc L, Sabatier L, et al. A cytogenetic study of 19 recurrent gliomas.
Cancer Genet Cytogenet
1994
;
76
:
85
–92.
14
Kallioniemi A, Kallioniemi OP, Sudar D, et al. Comparative genomic hybridization for molecular cytogenetic analysis of solid tumors.
Science
1992
;
258
:
818
–21.
15
Muleris M, Almeida A, Dutrillaux AM, et al. Oncogene amplification in human gliomas: a molecular cytogenetic analysis.
Oncogene
1994
;
9
:
2717
–22.
16
Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma.
N Engl J Med
2005
;
352
:
997
–1003.
17
Patel M, McCully C, Godwin K, Balis FM. Plasma and cerebrospinal fluid pharmacokinetics of intravenous temozolomide in non-human primates.
J Neurooncol
2003
;
61
:
203
–7.
18
Ma J, Murphy M, O'Dwyer PJ, Berman E, Reed K, Gallo JM. Biochemical changes associated with a multidrug-resistant phenotype of a human glioma cell line with temozolomide-acquired resistance.
Biochem Pharmacol
2002
;
63
:
1219
–28.
19
Tews DS, Nissen A, Kulgen C, Gaumann AK. Drug resistance-associated factors in primary and secondary glioblastomas and their precursor tumors.
J Neurooncol
2000
;
50
:
227
–37.
20
Paz MF, Yaya-Tur R, Rojas-Marcos I, et al. CpG island hypermethylation of the DNA repair enzyme methyltransferase predicts response to temozolomide in primary gliomas.
Clin Cancer Res
2004
;
10
:
4933
–8.
21
Trivedi RN, Almeida KH, Fornsaglio JL, Schamus S, Sobol RW. The role of base excision repair in the sensitivity and resistance to temozolomide-mediated cell death.
Cancer Res
2005
;
65
:
6394
–400.
22
Calatozzolo C, Gelati M, Ciusani E, et al. Expression of drug resistance proteins Pgp, MRP1, MRP3, MRP5 and GST-pi in human glioma.
J Neurooncol
2005
;
74
:
113
–21.
23
Esteller M, Herman JG. Generating mutations but providing chemosensitivity: the role of O6-methylguanine DNA methyltransferase in human cancer.
Oncogene
2004
;
23
:
1
–8.
24
Watanabe T, Katayama Y, Komine C, et al. O6-Methylguanine-DNA methyltransferase methylation and TP53 mutation in malignant astrocytomas and their relationships with clinical course.
Int J Cancer
2005
;
113
:
581
–7.
25
Rolhion C, Penault-Llorca F, Kemeny JL, et al. O(6)-methylguanine-DNA methyltransferase gene (MGMT) expression in human glioblastomas in relation to patient characteristics and p53 accumulation.
Int J Cancer
1999
;
84
:
416
–20.
26
Hermisson M, and Weller M. NF-κB-independent actions of sulfasalazine dissociate the CD95L- and Apo2L/TRAIL-dependent death signaling pathways in human malignant glioma cells.
Cell Death Differ
2003
;
10
:
1078
–89.
27
Koschny R, Koschny T, Froster UG, Krupp W, Zuber MA. Comparative genomic hybridization in glioma: a meta-analysis of 509 cases.
Cancer Genet Cytogenet
2002
;
135
:
147
–59.
28
Duesberg P, Stindl R, Hehlmann R. Origin of multidrug resistance in cells with and without multidrug resistance genes: chromosome reassortments catalyzed by aneuploidy.
Proc Natl Acad Sci U S A
2001
;
98
:
11283
–8.
29
Umehara H, Nishii Y, Morishima M, et al. Effect of cisplatin treatment on speckled distribution of a serine/arginine-rich nuclear protein CROP/Luc7A.
Biochem Biophys Res Commun
2003
;
301
:
324
–9.
30
Bergamaschi D, Samuels Y, O'Neil NJ, et al. iASPP oncoprotein is a key inhibitor of p53 conserved from worm to human.
Nat Genet
2003
;
33
:
162
–7.
31
Drivdahl R, Haugk KH, Sprenger CC, Nelson PS, Tennant MK, Plymate SR. Suppression of growth and tumorigenicity in the prostate tumor cell line M12 by overexpression of the transcription factor SOX9.
Oncogene
2004
;
23
:
4584
–93.
32
Ishii H, Vecchione A, Murakumo Y, et al. FEZ1/LZTS1 gene at 8p22 suppresses cancer cell growth and regulates mitosis.
Proc Natl Acad Sci U S A
2001
;
98
:
10374
–9.
33
Huang R, Lin Y, Wang CC, et al. Connexin 43 suppresses human glioblastoma cell growth by down-regulation of monocyte chemotactic protein 1, as discovered using protein array technology.
Cancer Res
2002
;
62
:
2806
–12.
34
Szeto CC, Chow KM, Lai KB, Szeto CY, Kwan BC, Li PK. Connective tissue growth factor is responsible for transforming growth factor-β-induced peritoneal mesothelial cell apoptosis.
Nephron Exp Nephrol
2006
;
103
:
166
–74.
35
Bhattacharya B, Miura T, Brandenberger R, et al. Gene expression in human embryonic stem cell lines: unique molecular signature.
Blood
2004
;
103
:
2956
–64.
36
Mukobata S, Hibino T, Sugiyama A, et al. M6a acts as a nerve growth factor-gated Ca(2+) channel in neuronal differentiation.
Biochem Biophys Res Commun
2002
;
297
:
722
–8.
37
Albig AR, Schiemann WP. Fibulin-5 antagonizes vascular endothelial growth factor (VEGF) signaling and angiogenic sprouting by endothelial cells.
DNA Cell Biol
2004
;
23
:
367
–79.
38
Merrill RA, See AW, Wertheim ML, Clagett-Dame M. Crk-associated substrate (Cas) family member, NEDD9, is regulated in human neuroblastoma cells and in the embryonic hindbrain by all-trans retinoic acid.
Dev Dyn
2004
;
231
:
564
–75.
39
Wine-Lee L, Ahn KJ, Richardson RD, Mishina Y, Lyons KM, Crenshaw EB 3rd. Signaling through BMP type 1 receptors is required for development of interneuron cell types in the dorsal spinal cord.
Development
2004
;
131
:
5393
–403.
40
Filmus J, Capurro M. Glypican-3 and α-fetoprotein as diagnostic tests for hepatocellular carcinoma.
Mol Diagn
2004
;
8
:
207
–12.
41
Kudo C, Ajioka I, Hirata Y, Nakajima K. Expression profiles of EphA3 at both the RNA and protein level in the developing mammalian forebrain.
J Comp Neurol
2005
;
487
:
255
–69.
42
Nagata T, Takahashi Y, Ishii Y, et al. Profiling of genes differentially expressed between fetal liver and postnatal liver using high-density oligonucleotide DNA array.
Int J Mol Med
2003
;
11
:
713
–21.
43
Zhang QH, Ye M, Wu XY, et al. Cloning and functional analysis of cDNAs with open reading frames for 300 previously undefined genes expressed in CD34+ hematopoietic stem/progenitor cells.
Genome Res
2000
;
10
:
1546
–60.
44
Geiman TM, Durum SK, Muegge K. Characterization of gene expression, genomic structure, and chromosomal localization of Hells (Lsh).
Genomics
1998
;
54
:
477
–83.
45
Yano M, Ouchida M, Shigematsu H, et al. Tumor-specific exon creation of the HELLS/SMARCA6 gene in non-small cell lung cancer.
Int J Cancer
2004
;
112
:
8
–13.
46
Stanchina L, Baral V, Robert F, et al. Interactions between Sox10, Edn3 and Ednrb during enteric nervous system and melanocyte development.
Dev Biol
2006
;
295
:
232
–49.
47
Colin C, Baeza N, Bartoli C, et al. Identification of genes differentially expressed in glioblastoma versus pilocytic astrocytoma using Suppression Subtractive Hybridization.
Oncogene
2006
;
25
:
2818
–26.
48
Galceran J, Sustmann C, Hsu SC, Folberth S, Grosschedl R. LEF1-mediated regulation of Δ-like1 links Wnt and Notch signaling in somitogenesis.
Genes Dev
2004
;
18
:
2718
–23.
49
Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma.
Cancer Res
2004
;
64
:
7011
–21.
50
Pitman M, Emery B, Binder M, Wang S, Butzkueven H, Kilpatrick TJ. LIF receptor signaling modulates neural stem cell renewal.
Mol Cell Neurosci
2004
;
27
:
255
–66.
51
Kanda Y, Mitani K, Kurokawa M, Yamagata T, Yazaki Y, Hirai H. Overexpression of the MEN/ELL protein, an RNA polymerase II elongation factor, results in transformation of Rat1 cells with dependence on the lysine-rich region.
J Biol Chem
1998
;
273
:
5248
–52.
52
Xie D, Yin D, Wang HJ, et al. Levels of expression of CYR61 and CTGF are prognostic for tumor progression and survival of individuals with gliomas.
Clin Cancer Res
2004
;
10
:
2072
–81.
53
Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells.
Nature
2004
;
432
:
396
–401.
54
Kondo T, Setoguchi T, Taga T. Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line.
Proc Natl Acad Sci U S A
2004
;
101
:
781
–6.
55
Dean RA, Talbot NJ, Ebbole DJ, et al. The genome sequence of the rice blast fungus Magnaporthe grisea.
Nature
2005
;
434
:
980
–6.