Dynamics of chemosensitivity and chromosomal instability in recurrent glioblastoma

S Spiegl-Kreinecker, C Pirker, C Marosi, J Buchroithner, J Pichler, R Silye, J Fischer, M Micksche, W Berger, S Spiegl-Kreinecker, C Pirker, C Marosi, J Buchroithner, J Pichler, R Silye, J Fischer, M Micksche, W Berger

Abstract

Glioblastoma multiforme is characterised by invasive growth and frequent recurrence. Here, we have analysed chromosomal changes in comparison to tumour cell aggressiveness and chemosensitivity of three cell lines established from a primary tumour and consecutive recurrences (BTL1 to BTL3) of a long-term surviving glioblastoma patient together with paraffin-embedded materials of five further cases with recurrent disease. Following surgery, the BTL patient progressed under irradiation/ lomustine but responded to temozolomide after re-operation to temozolomide. The primary tumour -derived BTL1 cells showed chromosomal imbalances typical of highly aggressive glioblastomas. Interestingly, BTL2 cells established from the first recurrence developed under therapy showed signs of enhanced chromosomal instability. In contrast, BTL3 cells from the second recurrence resembled a less aggressive subclone of the primary tumour. Although BTL2 cells exhibited a highly aggressive phenotype, BTL3 cells were characterised by reduced proliferative and migratory potential. Despite persistent methylation of the O6-methylguanine-DNA methyltransferase promoter, BTL3 cells exhibited the highest temozolomide sensitivity. A comparable situation was found in two out of five glioblastoma patients, both characterised by enhanced survival time, who also relapsed after surgery/chemotherapy with less aggressive recurrences. Taken together, our data suggest that pretreated glioblastoma patients may relapse with highly chemosensitive tumours confirming the feasibility of temozolomide treatment even in case of repeated recurrence.

Figures

Figure 1
Figure 1
CGH analysis profiles (for interpretation see Pirker et al, 2003) of (A) primary tumour-derived BTL1, (B) first recurrence-derived BTL2, and (C) second recurrence-derived BTL3 cells. Arrows in (A) indicate typical changes for glioblastoma multiforme. Asterisks in (B) indicate additional chromosomal aberrations in BTL2 as compared to BTL1. In (C) the asterisk indicates an aberration present also in BTL2 but not BTL1. Crosses in (C) represent changes unique to BTL3.
Figure 2
Figure 2
Cytogenetic analysis of the BTL glioblastoma cell model. Representative CGH (A), FISH (B, C), and CDD banding (DF) analyses of the indicated cell lines are shown. Arrows in (A) indicate typical changes for glioblastoma multiforme. For FISH analysis (B, C) a paint for whole chromosome 7 (red) and a BAC clone for the EGFR locus (green) were used. (D and E) show representative karyograms of the two subclones detected in BTL1.
Figure 3
Figure 3
In vitro growth dynamics of the BTL glioblastoma cell model. Growth curves for the indicated cell lines were established as described in Material and Methods. One of three experiments with comparable results is shown.
Figure 4
Figure 4
Migratory potential of the BTL glioblastoma cell model. Scratch assays were performed as described in Materials and Methods. (A) Representative photomicrographs taken at the indicated time points are shown. (B) To determine the relative closure of the gap, photomicrographs as shown in (A) were taken at different time points and measured using MetaMorph 6.1 software. The gaps’ widths immediately after wounding were arbitrarily set as 1. At least three experiments with comparable results were performed.
Figure 5
Figure 5
Antiproliferative and cytotoxic effects of temozolomide against BTL cells. (A) The indicated BTL cell lines were exposed to increasing temozolomide concentrations for 48 h and the rate of DNA synthesis was determined by 3H-thymidine incorporation. Data are given relatively to the untreated controls set as 1. Means of two experiments in triplicates are shown. (B) BTL cells were exposed for 5 days to the indicated temozolomide concentrations and the percentage of apoptotic/dead cells was determined by Hoechst 33258 and PI staining as described in Materials and Methods. One representative example out of three experiments delivering comparable results is shown.
Figure 6
Figure 6
Expression of chemoresistance-related genes in BTL glioblastoma cells. mRNA (A) and protein (B) levels encoded by the indicated chemoresistance genes were determined by RT–PCR and Western blot, respectively. (C) Methylation of the MGMT promoter was detected by methylation-specific PCR and a representative experiment is shown (m=methylated; u=unmethylated). Protein extracts of the glioblastoma cell lines GL80, GL54, and GL52 were used as positive controls for MGMT, MVP, and MRP1, respectively.

References

    1. Aghi M, Gaviani P, Henson JW, Batchelor TT, Louis DN, Barker II FG (2005) Magnetic resonance imaging characteristics predict epidermal growth factor receptor amplification status in glioblastoma. Clin Cancer Res 11: 8600–8605
    1. Albertson DG, Collins C, McCormick F, Gray JW (2003) Chromosome aberrations in solid tumors. Nat Genet 34: 369–376
    1. Aquilina G, Hess P, Branch P, MacGeoch C, Casciano I, Karran P, Bignami M (1994) A mismatch recognition defect in colon carcinoma confers DNA microsatellite instability and a mutator phenotype. Proc Natl Acad Sci USA 91: 8905–8909
    1. Berger W, Elbling L, Hauptmann E, Micksche M (1997) Expression of the multidrug resistance-associated protein (MRP) and chemoresistance of human non-small-cell lung cancer cells. Int J Cancer 73: 84–93
    1. Berger W, Elbling L, Micksche M (2000) Expression of the major vault protein LRP in human non-small-cell lung cancer cells: activation by short-term exposure to antineoplastic drugs. Int J Cancer 88: 293–300
    1. Berger W, Elbling L, Minai-Pour M, Vetterlein M, Pirker R, Kokoschka EM, Micksche M (1994) Intrinsic MDR-1 gene and P-glycoprotein expression in human melanoma cell lines. Int J Cancer 59: 717–723
    1. Berger W, Spiegl-Kreinecker S, Buchroithner J, Elbling L, Pirker C, Fischer J, Micksche M (2001) Overexpression of the human major vault protein in astrocytic brain tumor cells. Int J Cancer 94: 377–382
    1. Bocangel DB, Finkelstein S, Schold SC, Bhakat KK, Mitra S, Kokkinakis DM (2002) Multifaceted resistance of gliomas to temozolomide. Clin Cancer Res 8: 2725–2734
    1. Bonneau D, Longy M (2000) Mutations of the human PTEN gene. Hum Mutat 16: 109–122
    1. Bredel M (2001) Anticancer drug resistance in primary human brain tumors. Brain Res Brain Res Rev 35: 161–204
    1. Bredel M, Bredel C, Juric D, Duran GE, Yu RX, Harsh GR, Vogel H, Recht LD, Scheck AC, Sikic BI (2006) Tumor necrosis factor-alpha-induced protein 3 as a putative regulator of nuclear factor-kappaB-mediated resistance to O6-alkylating agents in human glioblastomas. J Clin Oncol 24: 274–287
    1. Buckner JC (2003) Factors influencing survival in high-grade gliomas. Semin Oncol 30: 10–14
    1. DeAngelis LM (2003) Benefits of adjuvant chemotherapy in high-grade gliomas. Semin Oncol 30: 15–18
    1. Duerr EM, Rollbrocker B, Hayashi Y, Peters N, Meyer-Puttlitz B, Louis DN, Schramm J, Wiestler OD, Parsons R, Eng C, von Deimling A (1998) PTEN mutations in gliomas and glioneuronal tumors. Oncogene 16: 2259–2264
    1. Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V, Baylin SB, Herman JG (2000) Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. New Engl J Med 343: 1350–1354
    1. Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG (1999) Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res 59: 793–797
    1. Etienne MC, Formento JL, Lebrun-Frenay C, Gioanni J, Chatel M, Paquis P, Bernard C, Courdi A, Bensadoun RJ, Pignol JP, Francoual M, Grellier P, Frenay M, Milano G (1998) Epidermal growth factor receptor and labeling index are independent prognostic factors in glial tumor outcome. Clin Cancer Res 4: 2383–2390
    1. Franceschi E, Omuro AM, Lassman AB, Demopoulos A, Nolan C, Abrey LE (2005) Salvage temozolomide for prior temozolomide responders. Cancer 104: 2473–2476
    1. Fujisawa H, Kurrer M, Reis RM, Yonekawa Y, Kleihues P, Ohgaki H (1999) Acquisition of the glioblastoma phenotype during astrocytoma progression is associated with loss of heterozygosity on 10q25-qter. Am J Pathol 155: 387–394
    1. Gerson SL (2002) Clinical relevance of MGMT in the treatment of cancer. J Clin Oncol 20: 2388–2399
    1. Grusch M, Fritzer-Szekeres M, Fuhrmann G, Rosenberger G, Luxbacher C, Elford HL, Smid K, Peters GJ, Szekeres T, Krupitza G (2001) Activation of caspases and induction of apoptosis by novel ribonucleotide reductase inhibitors amidox and didox. Exp Hematol 29: 623–632
    1. Harris LC, Margison GP (1993) Expression in mammalian cells of the Escherichia coliO6 alkylguanine-DNA-alkyltransferase gene ogt reduces the toxicity of alkylnitrosoureas. Br J Cancer 67: 1196–1202
    1. Hegi ME, Diserens AC, Godard S, Dietrich PY, Regli L, Ostermann S, Otten P, Van Melle G, de Tribolet N, Stupp R (2004) Clinical trial substantiates the predictive value of O-6-methylguanine-DNA methyltransferase promoter methylation in glioblastoma patients treated with temozolomide. Clin Cancer Res 10: 1871–1874
    1. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. New Engl J Med 352: 997–1003
    1. Hulsebos TJ, Troost D, Leenstra S (2004) Molecular-genetic characterisation of gliomas that recur as same grade or higher grade tumours. J Neurol Neurosurg Psychiatry 75: 723–726
    1. Inda MM, Fan X, Munoz J, Perot C, Fauvet D, Danglot G, Palacio A, Madero P, Zazpe I, Portillo E, Tunon T, Martinez-Penuela JM, Alfaro J, Eiras J, Bernheim A, Castresana JS (2003) Chromosomal abnormalities in human glioblastomas: gain in chromosome 7p correlating with loss in chromosome 10q. Mol Carcinog 36: 6–14
    1. Kang CS, Pu PY, Li YH, Zhang ZY, Qiu MZ, Huang Q, Wang GX (2005) An in vitro study on the suppressive effect of glioma cell growth induced by plasmid-based small interference RNA (siRNA) targeting human epidermal growth factor receptor. J Neurooncol 74: 267–273
    1. Lamszus K, Brockmann MA, Eckerich C, Bohlen P, May C, Mangold U, Fillbrandt R, Westphal M (2005) Inhibition of glioblastoma angiogenesis and invasion by combined treatments directed against vascular endothelial growth factor receptor-2, epidermal growth factor receptor, and vascular endothelial-cadherin. Clin Cancer Res 11: 4934–4940
    1. Leuraud P, Taillandier L, Medioni J, Aguirre-Cruz L, Criniere E, Marie Y, Kujas M, Golmard JL, Duprez A, Delattre JY, Sanson M, Poupon MF (2004) Distinct responses of xenografted gliomas to different alkylating agents are related to histology and genetic alterations. Cancer Res 64: 4648–4653
    1. Liu L, Markowitz S, Gerson SL (1996) Mismatch repair mutations override alkyltransferase in conferring resistance to temozolomide but not to 1,3-bis(2-chloroethyl)nitrosourea. Cancer Res 56: 5375–5379
    1. Liu W, James CD, Frederick L, Alderete BE, Jenkins RB (1997) PTEN/MMAC1 mutations and EGFR amplification in glioblastomas. Cancer Res 57: 5254–5257
    1. Mao X, Hamoudi RA (2000) Molecular and cytogenetic analysis of glioblastoma multiforme. Cancer Genet Cytogenet 122: 87–92
    1. Nagane M, Levitzki A, Gazit A, Cavenee WK, Huang HJ (1998) Drug resistance of human glioblastoma cells conferred by a tumor-specific mutant epidermal growth factor receptor through modulation of Bcl-XL and caspase-3-like proteases. Proc Natl Acad Sci USA 95: 5724–5729
    1. Nigro JM, Misra A, Zhang L, Smirnov I, Colman H, Griffin C, Ozburn N, Chen M, Pan E, Koul D, Yung WK, Feuerstein BG, Aldape KD (2005) Integrated array-comparative genomic hybridization and expression array profiles identify clinically relevant molecular subtypes of glioblastoma. Cancer Res 65: 1678–1686
    1. Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C, Schuler D, Probst-Hensch NM, Maiorka PC, Baeza N, Pisani P, Yonekawa Y, Yasargil MG, Lutolf UM, Kleihues P (2004) Genetic pathways to glioblastoma: a population-based study. Cancer Res 64: 6892–6899
    1. Pirker C, Holzmann K, Spiegl-Kreinecker S, Elbling L, Thallinger C, Pehamberger H, Micksche M, Berger W (2003) Chromosomal imbalances in primary and metastatic melanomas: over-representation of essential telomerase genes. Melanoma Res 13: 483–492
    1. Pirker C, Raidl M, Steiner E, Elbling L, Holzmann K, Spiegl-Kreinecker S, Aubele M, Grasl-Kraupp B, Marosi C, Micksche M, Berger W (2004) Whole genome amplification for CGH analysis: linker-adapter PCR as the method of choice for difficult and limited samples. Cytometry 61A: 26–34
    1. Rabik CA, Njoku MC, Dolan ME (2006) Inactivation of O6-alkylguanine DNA alkyltransferase as a means to enhance chemotherapy. Cancer Treat Rev 32: 261–276
    1. Rasheed BK, Stenzel TT, McLendon RE, Parsons R, Friedman AH, Friedman HS, Bigner DD, Bigner SH (1997) PTEN gene mutations are seen in high-grade but not in low-grade gliomas. Cancer Res 57: 4187–4190
    1. Saxena A, Shriml LM, Dean M, Ali IU (1999) Comparative molecular genetic profiles of anaplastic astrocytomas/glioblastomas multiforme and their subsequent recurrences. Oncogene 18: 1385–1390
    1. Schlegel J, Merdes A, Stumm G, Albert FK, Forsting M, Hynes N, Kiessling M (1994a) Amplification of the epidermal-growth-factor-receptor gene correlates with different growth behaviour in human glioblastoma. Int J Cancer 56: 72–77
    1. Schlegel J, Stumm G, Brandle K, Merdes A, Mechtersheimer G, Hynes NE, Kiessling M (1994b) Amplification and differential expression of members of the erbB-gene family in human glioblastoma. J Neurooncol 22: 201–207
    1. Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M, Ahr A, Blankenstein TJ, Kaufmann M, Diebold J, Arnholdt H, Muller P, Bischoff J, Harich D, Schlimok G, Riethmuller G, Eils R, Klein CA (2003) From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci USA 100: 7737–7742
    1. Scott CB, Scarantino C, Urtasun R, Movsas B, Jones CU, Simpson JR, Fischbach AJ, Curran Jr WJ (1998) Validation and predictive power of Radiation Therapy Oncology Group (RTOG) recursive partitioning analysis classes for malignant glioma patients: a report using RTOG 90–06. Int J Radiat Oncol Biol Phys 40: 51–55
    1. Shinojima N, Tada K, Shiraishi S, Kamiryo T, Kochi M, Nakamura H, Makino K, Saya H, Hirano H, Kuratsu J, Oka K, Ishimaru Y, Ushio Y (2003) Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res 63: 6962–6970
    1. Silber JR, Bobola MS, Ghatan S, Blank A, Kolstoe DD, Berger MS (1998) O6-methylguanine-DNA methyltransferase activity in adult gliomas: relation to patient and tumor characteristics. Cancer Res 58: 1068–1073
    1. Smith JS, Jenkins RB (2000) Genetic alterations in adult diffuse glioma: occurrence, significance, and prognostic implications. Front Biosci 5: D213–D231
    1. Spiegl-Kreinecker S, Buchroithner J, Elbling L, Steiner E, Wurm G, Bodenteich A, Fischer J, Micksche M, Berger W (2002) Expression and functional activity of the ABC-transporter proteins P-glycoprotein and multidrug-resistance protein 1 in human brain tumor cells and astrocytes. J Neurooncol 57: 27–36
    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New Engl J Med 352: 987–996
    1. Trivedi RN, Almeida KH, Fornsaglio JL, Schamus S, Sobol RW (2005) The role of base excision repair in the sensitivity and resistance to temozolomide-mediated cell death. Cancer Res 65: 6394–6400
    1. van den Bent MJ, Hegi ME, Stupp R (2006) Recent developments in the use of chemotherapy in brain tumours. Eur J Cancer 42: 582–588
    1. Wiltshire RN, Rasheed BK, Friedman HS, Friedman AH, Bigner SH (2000) Comparative genetic patterns of glioblastoma multiforme: potential diagnostic tool for tumor classification. Neuro-oncology 2: 164–173
    1. Zhu A, Shaeffer J, Leslie S, Kolm P, El-Mahdi AM (1996) Epidermal growth factor receptor: an independent predictor of survival in astrocytic tumors given definitive irradiation. Int J Radiat Oncol Biol Phys 34: 809–815
    1. Zhu Y, Parada LF (2002) The molecular and genetic basis of neurological tumours. Nat Rev Cancer 2: 616–626

Source: PubMed

3
S'abonner