the effects of irradiation on cell migration from glioblastoma multiforme biopsy spheroids

10
ORIGINAL PAPER Fabian Fehlauer Martina Muench Dirk Rades Lukas J. A. Stalpers Sieger Leenstra Paul van der Valk Ben Slotman Ernst J. Smid Peter Sminia Effects of irradiation and cisplatin on human glioma spheroids: inhibition of cell proliferation and cell migration Received: 1 March 2005 / Accepted: 21 June 2005 / Published online: 12 August 2005 Ó Springer-Verlag 2005 Abstract Purpose: Investigation of cell migration and proliferation of human glioma cell line spheroids (CLS) and evaluation of morphology, apoptosis, and immuno- histochemical expression of MIB-1, p53, and p21 of or- ganotypic muticellular spheroids (OMS) following cisplatin (CDDP) and irradiation (RT). Material and methods: Spheroids of the GaMg glioma cell line and OMS prepared from biopsy tissue of six glioblastoma patients were used. Radiochemosensitvity (5 lg/ml CDDP followed by RT) was determined using migration and proliferation assays on CLS. In OMS, histology and immunohistochemical studies of MIB-1, p53, and p21 expression were examined 24 and 48 h following treat- ment. Results: Combination treatment led to a migration inhibition of 38% (CDDP 13%; RT 27%) and specific growth delay of 2.6 (CDDP 1.3; RT 2.1) in CLS. Cell cycle analysis after combination treatment showed an accu- mulation of cells in the G2/M phase. In OMS, apoptosis increased, cell proliferation decreased, and p53/p21 expression increased more pronounced following CDDP+RT. No morphological damage was observed. Conclusion: CDDP can lead to enhancement of the RT effect in spheroids of both human glioma cell line spher- oids and biopsy spheroids from glioblastoma specimens. The exerted effect is additive rather than synergistic. Keywords Glioma Irradiation Cisplatin Spheroids Introduction Although radiotherapy following surgical resection is the most effective treatment for glioblastoma multiforme (GBM), the median survival in these patients is <1 year (Gonza`lez Gonza´lez and Hulshof 1993; Hulshof et al. 2001). The modest increase in survival time after radio- therapy treatment has been ascribed to the high intrinsic resistance of the gliomas to X-irradiation (Taghian et al. 1993, 1995). To improve survival in patients with malignant glial tumours new treatment strategies are required. Further approaches include chemotherapeutic agents as an adjuvant modality or as a radiosensitiser. Currently, several in vitro model systems to investi- gate the effects of irradiation and modulation of the radiation response of glioma are available. These include established cell lines, monolayer cultures of glioma lines, both early and late passage after initial isolation, and spheroids derived from cell lines, the so-called cell line spheroids (Carlson et al. 1983; Stuschke et al. 1993; Fehlauer et al. 2000). Another culture system used to study gliomas in vitro are the organotypic multicellular spheroids (OMS), which resemble the original architec- ture of the in vivo tumour (Darling et al. 1983; Bjerkvig et al. 1990; Fehlauer et al. 2004). It is assumed that spheroid cultures of both, cell lines and fresh tumour specimens, can better predict the in vivo response than monolayer cultures, since cell-cell contact, variation in cell cycle, altered metabolism, and diffusion of nutrients, oxygen or drugs may influence the outcome (Sutherland F. Fehlauer (&) M. Muench D. Rades Department of Radiation Oncology, Universita¨tsklinikum Eppendorf, University of Hamburg, Martinistr. 52, 20254 Hamburg, Germany E-mail: [email protected] Tel.: +49-40-428034031 Fax: +49-40-428032846 L. J. A. Stalpers F. Fehlauer Department of Radiation Oncology, AMC University of Amsterdam, Amsterdam, The Netherlands S. Leenstra Department of Neurosurgery, AMC University of Amsterdam, Amsterdam, The Netherlands P. van der Valk F. Fehlauer Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands B. Slotman E. J. Smid P. Sminia Department of Radiation Oncology, VU University Medical Center, Amsterdam, The Netherlands J Cancer Res Clin Oncol (2005) 131: 723–732 DOI 10.1007/s00432-005-0014-3

Upload: eur

Post on 21-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

ORIGINAL PAPER

Fabian Fehlauer Æ Martina Muench Æ Dirk Rades

Lukas J. A. Stalpers Æ Sieger LeenstraPaul van der Valk Æ Ben Slotman Æ Ernst J. Smid

Peter Sminia

Effects of irradiation and cisplatin on human glioma spheroids:inhibition of cell proliferation and cell migration

Received: 1 March 2005 / Accepted: 21 June 2005 / Published online: 12 August 2005� Springer-Verlag 2005

Abstract Purpose: Investigation of cell migration andproliferation of human glioma cell line spheroids (CLS)and evaluation of morphology, apoptosis, and immuno-histochemical expression of MIB-1, p53, and p21 of or-ganotypic muticellular spheroids (OMS) followingcisplatin (CDDP) and irradiation (RT). Material andmethods: Spheroids of the GaMg glioma cell line andOMS prepared from biopsy tissue of six glioblastomapatients were used. Radiochemosensitvity (5 lg/mlCDDP followed by RT) was determined using migrationand proliferation assays on CLS. In OMS, histology andimmunohistochemical studies of MIB-1, p53, and p21expression were examined 24 and 48 h following treat-ment. Results: Combination treatment led to a migrationinhibition of 38% (CDDP 13%; RT 27%) and specificgrowth delay of 2.6 (CDDP1.3; RT 2.1) inCLS. Cell cycleanalysis after combination treatment showed an accu-mulation of cells in the G2/M phase. In OMS, apoptosis

increased, cell proliferation decreased, and p53/p21expression increased more pronounced followingCDDP+RT. No morphological damage was observed.Conclusion: CDDP can lead to enhancement of the RTeffect in spheroids of both human glioma cell line spher-oids and biopsy spheroids from glioblastoma specimens.The exerted effect is additive rather than synergistic.

Keywords Glioma Æ Irradiation Æ Cisplatin Æ Spheroids

Introduction

Although radiotherapy following surgical resection is themost effective treatment for glioblastoma multiforme(GBM), the median survival in these patients is <1 year(Gonzalez Gonzalez and Hulshof 1993; Hulshof et al.2001). The modest increase in survival time after radio-therapy treatment has been ascribed to the high intrinsicresistance of the gliomas to X-irradiation (Taghian et al.1993, 1995). To improve survival in patients withmalignant glial tumours new treatment strategies arerequired. Further approaches include chemotherapeuticagents as an adjuvant modality or as a radiosensitiser.

Currently, several in vitro model systems to investi-gate the effects of irradiation and modulation of theradiation response of glioma are available. These includeestablished cell lines, monolayer cultures of glioma lines,both early and late passage after initial isolation, andspheroids derived from cell lines, the so-called cell linespheroids (Carlson et al. 1983; Stuschke et al. 1993;Fehlauer et al. 2000). Another culture system used tostudy gliomas in vitro are the organotypic multicellularspheroids (OMS), which resemble the original architec-ture of the in vivo tumour (Darling et al. 1983; Bjerkviget al. 1990; Fehlauer et al. 2004). It is assumed thatspheroid cultures of both, cell lines and fresh tumourspecimens, can better predict the in vivo response thanmonolayer cultures, since cell-cell contact, variation incell cycle, altered metabolism, and diffusion of nutrients,oxygen or drugs may influence the outcome (Sutherland

F. Fehlauer (&) Æ M. Muench Æ D. RadesDepartment of Radiation Oncology,Universitatsklinikum Eppendorf,University of Hamburg, Martinistr. 52,20254 Hamburg, GermanyE-mail: [email protected].: +49-40-428034031Fax: +49-40-428032846

L. J. A. Stalpers Æ F. FehlauerDepartment of Radiation Oncology,AMC University of Amsterdam, Amsterdam,The Netherlands

S. LeenstraDepartment of Neurosurgery,AMC University of Amsterdam, Amsterdam,The Netherlands

P. van der Valk Æ F. FehlauerDepartment of Pathology,VU University Medical Center, Amsterdam,The Netherlands

B. Slotman Æ E. J. Smid Æ P. SminiaDepartment of Radiation Oncology,VU University Medical Center, Amsterdam,The Netherlands

J Cancer Res Clin Oncol (2005) 131: 723–732DOI 10.1007/s00432-005-0014-3

1988; Kunz-Schunghart et al. 1998; Santini et al. 1999,2003; Walenta et al. 2000). The advantage of cell linespheroids is that they are relatively easy to obtain and tomaintain in culture. Treatment related changes of thegrowth kinetic and the outgrowth of tumour cells areestablished endpoints (Mahesparan et al. 1999; Gliem-roth et al. 2003; Kleynen et al. 2003). The advantage ofOMS over conventional spheroid culture is a remarkablestability in characteristics of the individual tumour:cellular heterogeneity (tumour cells, endothelial cell, andconnective tissue), differentiation pattern, proliferationrate and tumour cell ploidy are preserved. Individualtreatment related changes in terms of the histology andexpression of various proteins can by monitored withhistopathological and immunohistochemical techniques(Kaaijk et al. 1997).

Cisplatin (CDDP) is one of the most effective of allchemotherapeutic agents in solid tumours and hasshown to potentiate the effect of RT in various tumoursystems.

Mechanistically, CDDP molecules react with neucle-ophilic sites into DNA by forming adducts (crosslink-ing). In terms of cell cycle arrest, CDDP arrest cell cycleprogression in G1 and G2 phases, but it appears to beless active in inducing a p53 dependent G1 arrest. Cellswith disrupted p53 function tended to be less sensitive toRT (Haas-Kogan et al. 1999) and CDDP (O‘Connor1996) than cells with wt p53.

However, the combination of RT and CDDP showedunsatisfactory clinical results in malignant gliomas(Anonymus 1991), which might be due to interpatientvariation in radiochemosensitivity, intertumoural heter-ogeneity, and other physiological conditions. Localchemotherapy with CDDP-depot for GBM in combi-nation with RT was suggested to be an more effectiveapproach for those patients (Sheleg et al. 2002).

A number of in vitro studies have been publishedconcerning radiochemosensitivity of glioma cell lines byCDDP (Wilkens et al. 1996; Wolff et al. 1998, 1999;Yount et al. 1998; Fehlauer et al. 2000; Reichert et al.2002). CDDP in combination with RT exerted anadditive effect on early passage glioma cell cultures asshown by the unaltered value of the a-parameter forreproductive cell death of the linear quadratic model(Fehlauer et al. 2000).

In the present study the effect of CDDP and RT wasevaluated on spheroids from the human GaMg cell lineas well as on OMS derived from fresh tumour tissue ofnine GBM patients.

Methods

Monolayer cell culture

The GaMg cell line was obtained from a 42-years oldfemale with histologically proven GBM (Akslen et al.1988). GaMg cells were cultured as monolayer in Dul-becco’s modified Eagles medium (DMEM, Gibco BRL,

UK) supplemented with 10% fetal calf serum, 2 mML-glutamine and 50 lg/ml gentamycine (Gibco BRL) in25 cm2 culture flasks in a 37�C, 5% CO2, 95% humidi-fied air incubator.

Cell line spheroids

Spheroid cultures of GaMg were formed by liquid-overlay technique (Carlsson et al. 1983). In short,exponentially growing monolayers were trypsinized and5·106 cells were seeded in 20 ml of growth medium into80-cm2 agar-coated tissue culture flasks. After 10 days inculture, spheroids with diameter between 200 and250 lm were selected for the experiments. These spher-oids do not present central necrosis and have very fewhypoxic cells (Mueller-Klieser et al. 1986; Sutherland1986; Sminia et al. 2003).

Organotypic multicellular spheroids (OMS)

Fresh tumour tissue was obtained at surgery (openresection) from nine patients with a primary glioblas-toma, classified according to the World Health Organi-zation (WHO). The specimens were taken in tumourareas corresponding to regions with contrast enhance-ment on preoperative computerised tomography scansand transferred in Dulbecco’s modification of Eagle’smedium (DMEM; Flow Laboratories, Scotland).

For preparation of the spheroids, glioma tissue wasprocessed in the laboratory within 2 h from resection:representative tumour tissue was selected, based on botha preliminary frozen section histopathological diagnosismade by the responsible neuropathologist and on themacroscopic appearance. Blood and necrotic tissue wereremoved from tumour specimens and fragments of 0.5–1 mm3 were dissected with sterile 21G2 0.8·50 needles.48-well plates were coated with 100ll of a semisolidagarose gel, consisting of a 1:1 mixture of 1.5% agaroseand of DMEM supplemented with 4 mM L-glutamine,100 lg/ml gentamicin (all from Gibco BRL, U.K.) and20% heat-inactivated human serum (Bio-Whittaker,MD, USA). After the agarose had gelled, 300ll of cul-ture medium consisting of DMEM supplemented with2 mM L-glutamine, 100 lg/ml gentamicin and 10%heat-inactivated human serum was added to each well.

One biopsy fragment was transferred to each well ofthe 48-well plates. The spheroids were kept in a standardtissue culture incubator (98% humidity, 5% CO2, 37�C)and the culture medium was changed once a week.Cultured tissue fragments were studied twice a weekwith a phase-contrast microscope and only fragmentsthat became spherical within the first week were used forfurther study. Only tissue from patients with a fractionof fragments that formed spheroids of more than 40%were included in this study. Two weeks after onset of thecultures, the biopsy spheroids were used in the experi-ments.

724

Radiation and cisplatin treatment

Irradiation set up

Speroids were single dose irradiated (OMS: 20 Gy; CLS:10 Gy) using an orthovolt X-ray generator operating at250 kV and 15 mA, with a 0.5 mm thick Cu filter and atube opening of 8·8 cm. Spheroids were irradiated inpetri dishes (60 mm), containing 3 ml of culture mediumat a temperature of 37�C. Former experiments revealedthis dose range to be effective without sterilising thespheroids (Kleynen et al. 2003; Fehlauer et al. 2004).

Cisplatin treatment set up

Cisplatin (cis-diammine dichloroplatinum, CDDP;Platosin, Pharmachemie, Haarlem, NL) concentrationused was 5lg/ml, which is known from former experi-ments to be well incorporated by spheroids withoutpermitting growth (data not shown). For incubationwith CDDP, the spheroids were transferred into a60 mm petri dish. Incubation duration was 4 h, so therewas no need to use agar coated dishes, as this is too shorta period of time for the spheroids to attach to the bot-tom. After incubation the spheroids were washed twotimes with PBS and once with DMEM. Immediatelythereafter, the spheroids were irradiated.

Treatments

Thirty-six spheroids from each tumour were treated. Anequal number of spheroids from each tumour was shamtreated: At 24 and 48 h following treatment (sham, RT,CDDP, CDDP+RT) six spheroids from each groupwere fixed in 4% neutral buffered formaldehyde solutionfollowing treatment. Subsequently, spheroids were pro-cessed and embedded in paraffin for histological andimmunohistochemical analysis.

Tumour cell migration (cell line spheroids)

Following treatment, spheroids were placed individuallyinto uncoated 16-mm multi-well dishes which were filedwith 1 ml DMEM. Each treatment group consisted ofsix spheroids. One group was used as control. Thespheroid plated within 2 h, and the cellular outgrowthfrom the spheroid was defined as a colony. Twoorthogonal colony diameters were measured daily over a4-day period (96 h). by phase contrast microscope, andthe migratory capacities of the glioma cells were thendetermined by calculation the colony areas from thediameter measurements.

Growth delay essay (cell line spheroids)

For experiments, GaMg spheroids were placed individ-ually into 16-mm multiwell dishes (Nunc), which were

base-coated with 0.5 ml 0.75% DMEM-agar and filledwith 1 ml of DMEM. Six spheroids were selected for thestudy groups (sham, CDDP, RT, CDDP+RT). Aftertreatment, the dishes were incubated at 37�C. Thediameters of the spheroids were measured two times aweek in a phase contrast microscope over a 15-dayperiod, and the spheroid volume was calculated (volumemeasurement 4/3 pr3).

Once a week the culture medium was changed. Inorder to measure growth delay, the average time re-quired to reach ten times the initial spheroid volume wasdetermined. Specific growth delay (SGD) values werecalculated by dividing growth delay of treated samplesby the corresponding doubling times of control samples.

Flow cytometric analysis of cell cycle distribution(cell line spheroids)

To analyse the cell cycle changes, cells were plated andtreated as described above. At 24 h after initial plating,10 lM bromodeoxyuridine (BrdU) was administeredfrom a 100· stock. After 2 h, the cells were harvested,fixated in 70% ethanol in phosphate buffered saline(PBS) and stored at �20�C until immunofluorescentstaining. Ethanol-fixated cells were centrifuged (1 min,2,200 rpm), resuspended in 1 ml pepsin solution(0.4 mg/ml 0.1 N HCl) and incubated for 30 min atroom temperature. Subsequently, the DNA was dena-tured by a 30 min incubation in 1 ml 2 N HCl at 37�C.After washing with PBTb (PBS, Tween-20 0.05% v/v,bovine serum albumin (Sigma) 20 mg/ml, pH 7.4, thepellet was resuspended in 0.1 ml rat anti-BrdU (HarlanSeralab, Loughborough, UK, diluted 1:100 in PBTb),and incubated at room temperature for 30 min. Afterwashing with PBTg (PBS, Tween-20 0.005 v/v, normalgoat serum (Dako, Glostrup, Denmark) 1% v/v,pH 7.4), the pellet was resuspended in 0.1 fluoresceinconjugated goat-anti-rat IgG (Jackson, nr 112–015–102,West Grove, Pa., USA, diluted 1:100 in PBTg) andincubated at room temperature in the dark for 30 min.Propidium-iodine and ethanol were added to an end-concentration of 1 lg/ml and 30% respectively. Sampleswere stored at 4�C until flow cytometric analysis. Sam-ples were syringed through a 21-gauge needle to reducecell aggregation before flow cytometry (FACScan cy-tometer, Becton Dickenson, San Jose, CA, USA). Thedistribution of cells over the cell cycle was analysed withWindows Multiple Document Interface Flow CytometryApplication (WinMDI) by placing windows around G0/G1, S and G2/M populations.

Morphology and apoptosis (organotypic multicellularspheroids)

Paraffin sections (5 lm) of spheroids were placed onorganosilan-coated (3-aminopropyltriethoxysilane; Sig-ma, USA) object slides and dried overnight at 37�C.

725

Paraffin sections of the original tumour were similarlyprocessed. Histology of the spheroids was evaluated inHaematoxylin–Eosin stained sections; the same sectionswere also used for the assessment of apoptosis. Clustersof intensely basophilic nuclear fragments or an intenselybasophilic, shrunk nucleus were construed as morpho-logic evidence of the apoptotic process (Kerr et al.1972).

Immunohistochemistry (OMS)

Monoclonal antibodies were used for immunohisto-chemistry (MIB-1, p53, p21), using the Avidin-Biotin-peroxidase Complex (ABC-) technique according to themanufacturer’s protocol (MIB1, Ki67 protein, Immu-notech; DO7, wild type and mutant p53, DAKO; p21, p21 protein, PharMingen).

In brief, after sections (5lm) had undergo dewaxingand rehydration, endogenous peroxidase activity wasblocked for 30 min in methanol containing 0.3%hydrogen peroxide. Antigen retrieval was performed forall monoclonal antibodies by incubating slides in a cit-rate buffer solution (2.94 mg/ml trisodium citrate dihy-drate in distilled water; pH=6) in a microwave oven at180�C for 15 min. Thereafter, the slides were preincu-bated with normal rabbit serum in a 1:50 dilution inphosphate buffered saline with 1% bovine serum albu-mine (PBS/BSA) for 10 minutes. Subsequently, sectionswere incubated with monoclonal antibodies (overnight,4�C), and incubated with biotin-conjugated rabbit anti-mouse immunoglobulin or biotin-conjugated swine anti-rabbit immunoglobulin (all Dako, Denmark). Afterincubation with streptavidin-biotinylated horseradishperoxidase complex (dilution 1:200; Dako Denmark),peroxidase activity was developed using 3,3-diam-inobenzidine-tetrachloride (Sigma, USA) in 0.1%hydrogen peroxide as a chromogen. All sections werelightly counterstained (20 s) with haematoxylin, dehy-drated, and mounted. The signal for p21 was enhancedby the catalysed reporter deposition technique. Positivecontrol sections from known immunopositive tumourswere included for all antibodies. Negative controlsconsisted of sections of each group whereby the primaryantibody step was omitted.

Assessment and image analysis (OMS)

The evaluation of apoptosis and for all antibodies wasassessed with a conventional light microscope. Theapoptotic index was defined as percentage of apoptotictumour cells, identified as ‘apoptotic bodies’ (Kerr et al.1972). For p53, p21 and proliferation activity (MIB-1),all identifiable nuclear staining were regarded as posi-tive. The labelling index was defined as the percentage ofimmunopositive tumour cells in the spheroids. Noendothelial or haematogenous cells were included in thecounts.

The multiple factor of protein expression if treatmentgroups compared to the controls was calculated for eachtreatment group: a difference of a multiple factor >1was defined as a marked treatment effect.

The relative apoptosis count and p53, p21 and MIB-1positivity of the spheroids from each tumour was ex-pressed as % of the spheroids with the maximum posi-tivity (100%) from that tumour. Statistical comparisonof the data was performed using a unpaired sample t-testafter analysis of variance. P values <0.05 were con-sidered as significant.

Results

Cell line Spheroids

Tumour cell migration

The directorial cell migration from GaMg spheroids wasdetermined for treated and untreated spheroids (Fig. 1).,The outgrowth area was reduced by 38% (RT), 13%(CDDP), and 27% (CDDP+RT) as evaluated onday—following treatment. Combination therapy led to asignificant additional inhibition of cell migration(P<0.05).

Spheroid volume

In Fig. 2 shows the effect of irradiation alone or com-bined with cisplatin on GaMg spheroids. Combinationtreatment induced an additional growth delay. A sig-nificant reduction in spheroid growth was observed forCDDP+RT of 71% (specific growth delay, SGD 2.6),for RT alone of 42% (SGD 2.1), and for CDDP alone of32% (SGD 1.3) on day 20 (P<0.05).

0

2

4

6

8

10

12

14

16

18

0 2

days

area

(m

m2)

ControlCDDPRTCDDP+RT

1 3 4

Fig. 1 Tumour cell migration for GaMg cell line spheroidsfollowing RT, CDDP, and CDDP+RT. Each treatment groupconsisted of six spheroids. Changes at day 4 were significant(P<0.05)

726

Cell cycle distribution

After RT alone and in combination with CDDP, theflow cytometic DNA histograms reveals an more pro-nounced accumulation of GaMg cells in the G2/M phaseof the cell cycle (55, 59% respectively). The percentagesof the cells in G1, S, G2/M for (1) control, (2) RT, (3)CDDP, and (4) combination of both were (1) 45%,28%, 27%, (2) 27%, 18%, 55%, (3) 37%, 21%, 42%,and (4) 24%, 17%, 59%. Cell distribution of the treat-ment groups was not statistically significant.

Organotypic multicellular spheroids

Spheroid culture and volume measurements

Organotypic multicellular spheroids established from sixof the nine tumour samples could be included in thisstudy (S13, S14, S17, S18, S19, and S21). Phase contrastmicroscopy showed that approximately 70% of all tu-mour fragments prepared from fresh tissue formedspheroids within 1 week of culture. Fragments of threepatients could not be used for further experiments be-cause single cells and clumps started to be shed from thefragments without spheroid formation (S15, S16, S20).

During the culture period and following treatment,the volume of the spheroids from four tumours main-tained steady. Spheroids from two tumours showed adecrease of volume, more pronounced 48 h after com-bination treatment but not statistically significant fromcontrols.

Morphology and apoptosis

Histologically the spheroids closely resemble the originalindividual tumour. As in the tumour in vivo, OMScontained tumour cells, endothelial cells and connective

tissue. Typical features of glioblastoma were observed inuntreated and treated spheroids, including increasedcellularity, nuclear atypia, mitotic figures and non-functional capillaries, of which a few showed vascularproliferation. Neither necrosis, nor perinecrotic pseud-opalisading cellular components were observed.

Compared to the control spheroids, in none of thetreated spheroids a major morphological damage couldbe observed up to 48 h after treatment. No decrease ofcell density or changes in the glial structure was seen.Capillaries were preserved in both control and treatedspheroids of both follow-up intervals.

The number of apoptotic tumour cell increased inspheroids 24 and 48 h after treatment. Apoptosis,identified as clusters of intensely basophilic nuclearfragments were seen in both irradiated and controlspheroids from all groups.

Following CDDP and RT, individual tumoursshowed in five of six cases an maximal enhancement ofapoptotic bodies at both time points, were as a markedeffect was seen in three of six cases (S17, S18, S21) at the24 h interval, and two of six cases (S17, S18) at the 48 hinterval (Figs. 3, 4). CDDP alone was the most effectivetreatment modality in one of six cases (24 h: S13; 48 h:S19), were as the changes were only slightly more pro-nounced if compared to the other treatment groups.

Overall, a significant increase in apoptosis was de-tected following RT alone (P=0.04) and CDDP+RTtreatment (P=0.01, Table 1).

Cell proliferation

MIB-1 immunopositivity was exclusively nuclear. Con-trol spheroids showed a remarkable wide range of MIB-1 tumour cells (3–28%) which were mostly centred to-ward the peripheral zone. The mean labelling index andthe relative proliferation index was lower in spheroids oftreated groups compared to control (Fig. 3, Table 1).

Following CDDP and RT, individual tumoursshowed 24 h later in all cases an maximal decreasedproliferation labelling index, while a marked effect wasseen in two cases (S18, S21). At the 48 h interval, com-bination treatment let to decreased MIB-1 labelling in-dex in three of six cases, were as a marked effect wasonly seen in spheroids of tumour S21 (Fig. 4). CDDPalone was the most effective treatment modality in oneof six cases (48 h: S18).

Overall, CDDP+RT treated spheroids showed thehighest decrease of MIB-1 index, but changes were notsignificant.

p53 expression

In the resection material from the original glioblastomasthe overall p53 expression was less than 5%. In spher-oids established from these tumours, the absolute per-centage ranged between 2 and 8% and immunopositivitywas exclusively nuclear. A wide variety in the percentage

0

0,1

0,2

0,3

0,4

0,5

0,6

0,7

0 5 10 15 20

days

volu

me

in m

m3

ControlCDDPRTCDDP+RT

Fig. 2 Volume growth of GaMg cell line spheroids following RT,CDDP, and CDDP+RT. Each treatment group consisted of sixspheroids. Changes at day 20 were significant (P<0.05)

727

of p53 immunoreactive tumour cells was documentedbetween spheroids within the same group.

Following CDDP and RT, individual tumoursshowed in four of six cases an maximal enhancementof p53 expression at both time points, were as amarked effect was seen in one of six cases (S18) at the24 h interval, and two of six cases (S17, S18) at the

48 h interval (Figs. 3, 4). P53 expression was highestfollowing RT alone in one of six cases (24 h: S13;48 h: S14), were as the changes were only slightly morepronounced if compared to the other treatmentgroups.

Overall, a significant increase in p53 expression wasfound following RT alone (P=0.012), CDDP alone

Fig. 3 Apoptosis andimmunohistochemicalexpression of organotypicmulticellular spheroids (OMS)of six patients (S13, S14, S17,S18, S19, S21) as determined24 h (left column) and 48 h(right column) following RT,CDDP, and CDDP+RTcompared to the untreatedcontrols. From top, relativeapoptotic index, relativeexpression of p53, p21, andrelative cell proliferation(+, P<0.05)

728

(P=0.008) and CDDP+RT treatment (P=0.008,Table 1).

p21 expression

All original tumours contained less than 5% p21immunoreactive tumour cells. In untreated spheroids,the mean p21 expression was 1–2%.

Following CDDP and RT, individual tumoursshowed in two of six cases an maximal enhancement ofp21 expression at 24 h interval (S17, S18), and three ofsix cases at the 48 h interval (S14, S18, S21). In all ofthese cases a marked effect was detected. RT alone wasthe most effective modality in one of six cases with amarked effect 24 h following treatment (S14, Fig. 3).

Overall, p21 protein expression increased significantly(P<0.05) at 24 h following RT alone and CDDP+RTtreatment (Table 1).

Discussion

The present work shows that cisplatin (CDDP) andradiotherapy (RT) exert an additive effect in humanglioma spheroids, established from the human GaMgcell line grown as cell line spheroids and from fresh tu-mour tissue of six glioblastoma patients grown as OMS.

At present, the parallel utilisation of these two assaysystems was not used in experimental neuro-oncologyfor radiochemotherapy testing. OMS derived from hu-

Fig. 4 a Haematoxylin–Eosinstained section of a organotypicmuticellular spheroid (OMS). bImmunohistochemical stainingof p53. OMS showed asignificant increased rate ofapoptosis (a) and p53 proteinexpression (b) 24 h afterirradiation and cisplatintreatment. Originalmagnification, ·40. c and dImmunohistochemical stainingof Ki-67 (MIB-1). The numberof proliferating cells was higherin control (c) compared tocorresponding singe modalitytreated spheroids or to thecorresponding spheroidstreated with irradiation andcispaltin (d). Originalmagnification, ·10

Table 1 Mean labelling index(range) of apoptosis, p53, p21and MIB-1 protein expressionin OMS (n=6) at 24 and 48 hfollowing RT, CDDP, andCDDP+RT

NS Not significant

Follow up (h) Control RT CDDP CDDP+RT

Apoptosis 24 1.3±0.4 1.8±0.7 2.3±1.7 2.9±2.0P value P=0.01 NS P=0.04

48 1.6 ± 1.0 1.8 ± 0.9 2.4 ± 1.3 2.9 ± 1.3P value NS P=0.04 P=0.022p53 24 6.8±3.6 26.4±17.8 18.6±10.8 30.0±19.0P value P=0.012 P=0.008 P=0.008

48 5.2±2.9 22.0±28.7 12.8±10.2 30.5±27.5P value NS NS NSp21 24 1.1±0.4 4.8±2.2 3.6±3.2 7.8±4.7P value P=0.001 NS P=0.006

48 1.3±0.8 9.5±7.91 2.7±1.4 11.0±14.4P value NS NS NSMIB-1 24 4.8±2.0 4.4±2.9 3.7±2.6 3.3±2.1P value NS NS NS

48 4.3±1.5 3.3±2.6 4.2±3.9 3.3±3.3P value NS NS NS

729

man tumour tissue represents a methodically and bio-logically different three-dimensional culture model,which reflects in vivo situation much more reliable thancell lines or cell line spheroids do.

Spheroids derived from cell lines lack the cellularheterogeneity of OMS as they are grown from a singleimmortal cell line. However, several studies have shownthat these spheroids still have some characteristics of theoriginal tumour, which include aspects of tumour mor-phology and behaviour. In cell line spheroids, as inOMS, cell–cell contact, variation in cell cycle distribu-tion, diffusion effects, altered metabolism, and hypoxiaare presumed to influence the outcome of radiationtreatment (Olive and Durand 1994; Santini 1999; Sminiaet al. 2003). When these spheroids grow, the number ofproliferative cells decreases and the number of quiescentcells increases. Proliferation becomes limited to the outerrim of the cell line spheroids. Growth delay measure andoutgrowth assay following treatment were establishedendpoints and represent the function of surviving cells,re-growth rate, and migration property which reflectsboth radiocurability and radiosensitivity of experimentalglioma.

For human glioblastomas (GBM), the individualtreatment response of RT and radiosensitization byCDDP can be monitored excellent with OMS due to thefact, that the original tumour cell architecture, geneticprofile, extracellular matrix component, connective tis-sue, vessels and macrophages were preserved underexperimental conditions. The volume of these spheroidsmaintain stable in terms of growth, which can beattributed to a balance between cell proliferation andcell shedding. Conventional morphological studiesincluding immunohistochemical analysis of variousproteins following treatment are suitable endpoints(Bjerkvic et al. 1990; Kaaijk et al. 1995; Fehlauer et al.2004). However, cell line spheroids have the advantagethat they are relatively easy to obtain and to maintain inculture compared to OMS.

Most in vitro data on the response of GBM to RTand CDDP are based on monolayer cell cultures. In aprevious study, we evaluated the effect of RT andCDDP on primary, early passage cell cultures fromhuman GBM tumour specimens (Fehlauer et al. 2000).The survival fraction of cells following two Gray irra-diation (SF2) showed rather high values in clonogenicassay (0.61–0.72) demonstrating the high intrinsic ra-dioresistance of these cells. In combination with CDDP,an additive rather than radiosensitising effect could berecognised.

Although a CDDP-resistant human glioma cell linerequires a higher CDDP dose, the same level of sensiti-sation can be induced as in a sensitive glioma cell line,suggesting CDDP to be an effective additional cytotoxicagent to RT (Wilkens 1996).

The molecular mechanisms at the origin of theadditive effect of CDDP to RT is not yet fully under-stood. CDDP induces DNA adducts, which are gener-ally repaired by a recombination-like pathway. In

parallel, ionising radiation produces double strandbreaks, which are more likely recognised by the Kuprotein and rejoined by nonhomologous end joining,suggesting that CDDP treatment combined with radia-tion leads to the interplay of two major DNA repairpathways (Dewit 1987; Biston et al. 2004).

In our study, CDDP enhanced the effect of irradia-tion in both cell line spheroids and OMS additively. Theregrowth and migration capacity of GaMg cell linespheroids was inhibited in a additive manner whereassubpopulations of cell line spheroids survive and con-tinue to grow and migrate despite combination therapy.If compared with single mode treatment, combinationtherapy led to a reduction of the migration capacity offactor 0.8 (RT) and factor 0.7 (CDDP). A specificgrowth delay of 2.6 following combination therapycould be observed (CDDP 1.3; RT 2.1). The exertedeffect of both assays among CLS was additive ratherthan supra-additive or synergistic.

After irradiation and cisplatin, flow cytometric anal-ysis revealed only minor differences in cell cycle distri-butions if compared to single mode treatment.Radiotherapy alone and in combination with CDDPshowed an increased delay in G2 phase. The mechanismsfor the delay in the G2 phase have been demonstrated invirtually all eukaryotic cells examined in response toirradiation. After a single irradiation dose, a disturbancein DNA synthesis inflicting during late G1 or early Sphase is associated with subsequent accumulation ofcells in the G2-phase (Zatterstrom et al. 1995).

In OMS, combination treatment did not induce earlymajor histological damage or changes of the glialstructure up to 48 h post-treatment as evaluated by lightmicroscopy.

We found an remarkable increased apoptotic cell ratein spheroids of three of six patients following 20 Gyirradiation and 5 lg/ml CDDP, which was more pro-nounced 24 h post-treatment.

We also investigated the time-dependent expressionof p53 and p21, both proteins involved of control of cellcycle and apoptosis. Interestingly, we found a remark-able co-expression in spheroids S17 and S18. In thesecases, elevated p21 expression was detected following24 h following RT and CDDP, where as elevated p53expression was more pronounced at 48 h follow-up.

The evaluation of p21 expression, a downstreameffector of wt p53, has been widely used an indirect toolto assess for the functional status of p53 gene. A higherpercentage of p21 immunoreactive cells was docu-mented in p53 astrocytomas with wt p53 gene com-pared to either mutant p53 haboring p53immunonegative tumours. Thus, the expression of p21protein mediates p53-dependent cell cyclus arrest orapoptosis. It was demonstrated that the p21 gene isinduced by pathways dependent and independent fromthe functionally intact p53 tumour suppressor protein(Ono et al. 1997). The observed high variation of p53and p21 expression within the same groups underlinesthe heterogeneity of GBM. Furthermore, tumour cell

730

proliferation decreased additively in the combinationgroups in both assays.

Our results suggest that the limited therapeutic gainof RT in combination with CDDP, i.e., prolongation ofdisease-free interval and survival in GBM patients, canbe achieved due to a temporary cell cycle arrest, an earlyapoptotic response, decrease in proliferating tumourcells, and reduced migration potential. Clinically, theadditive treatment effect of RT and CDDP seems to bemore pronounced, when CDDP is administered locallyin GBM patients (Sheleg et al. 2002).

Finally, the combinational use of glioma cell linespheroids and organotypic multicellular spheroids de-rived from patients’ glioblastoma tissue are useful sup-plemental 3-dimensional assays for translationalresearch in neuro-oncology to study individual treat-ment effects of irradiation in combination with experi-mental therapeutic approaches on gliomas.

Acknowledgments The authors acknowledge Dr. R. Bjerkvig(Department of Anatomy and Cell Biology, University of Bergen,Norway) for his hospitality during the laboratory visit of Dr. F.Fehlauer in Bergen and his critical comments on the manuscript.We also wish to thank the members of the European Cancer Centre(ECC) Neuro-Oncology Group for their collaborations, insightfuldiscussions and encouragement. Dr. F. Fehlauer was research fel-low and supported by a grand of the Deutsche Forschungsgeme-inschaft (Bonn, Germany) and the ECC (Amsterdam, TheNetherlands). Furthermore, we thank Dr. Dr. A.J. Terzis(Department of Neurosurgery, University of Luebeck, Germany)for providing the GaMg cell line.

References

Akslen LA, Andersen KJ, Bjerkvig R (1988) Characteristics ofhuman and rat glioma cells grown in a defined medium. Anti-cancer Res 8:797–804

Anonymus (1991) Cisplatin does not enhance the effect of radiationtherapy in malignant gliomas. EORTC brain tumour group.Eur J Cancer 27:568–571

Biston MC, Joulbert A, Adam JF, Elleaume H, Bohic S, CharvetAM, Esteve F, Foray N, Balosso J (2004) Cure of Fisher ratsbearing radioresistant F98 glioma treated with cis-platinum andirradiated with monochromatic synchroton X-rays. Cancer Res64:2317–2323

Bjerkvig R, Tønnesen A, Laerum OD, Backlund EO (1990) Mul-ticellular tumor spheroids from human gliomas maintained inorgan culture. J Neurosurg 72:463–475

Carlsson J, Nilsson K, Wesermark B, Ponten J, Sundstrom C,Larsson E, Bergh J, Pahlmann S, Busch C, Collins VP (1983)Formation and growth of multicellular spheroids of humanorigin. Int J Cancer 31:523–533

Darling JL, Oktar N, Thomas DGT (1983) Multicellular tumourspheroids derived from human brain tumours. Cell Biol Int Rep7:23–30

Dewit L (1987) Combined treatment of radiation and cis-diamm-inedichloroplatinum (II): a review of experimental and clinicaldata. Int J Radiat Oncol Biol Phys 13:403–426

Durand RE, Olive PL (2001) Resistance of tumor cells tochemo- and radiotherapy modulated by the three-dimensionalarchitecture of solid tumors and spheroids. Methods Cell Biol4:211–233

Fehlauer F, Barten-Van Rijbroek AD, Stalpers LJ, Leenstra S,Lindeman J, Tjahja I, Troost D, Wolbers JG, van der Valk P,Sminia P (2000) Additive cytotoxic effect of cisplatin and

X-irradiation on human glioma cell cultures derived frombiopsy-tissue. J Cancer Res Clin Oncol 126:711–726

Fehlauer F, Stalpers LJ, Panayiotides J, Kaaijk P, GonzalezGonzalez D, Leenstra S, van der Valk P, Sminia P (2004) Effectof single dose irradiation on human glioblastoma spheroids invitro. Oncol Rep 11:477–485

Gliemroth J, Feyerabend T, Gerlach C, Arnold H, Terzisi AJA(2003) Proliferation, migration, and invasion of glioma cellsexposed to fractionated radiotherapy in vitro. Neursurg Rev26:198–205

Gonzalez Gonzalez D, Hulshof MC (1993) Radiotherapy. In:Twijnstra A, Keyser A, Ongerboer de Visser BW (eds) Neuro-oncology. Primary tumors and neurological complications ofcancer. Elsevier, Amsterdam, pp 134–158

Haas-Kogan DA, Kogan SS, Yount G, Hsu J, Haas H, Deen DF,Israel MA (1999) P53 function influences the effect of frac-tionated radiotherapy on glioblastoma tumors. Int J RadiatOncol Biol Phys 43:399–403

Hulshof MC, Koot RW, Schimmel EC, Dekker F, Bosch DA,Gonzalez Gonzalez D (2001) Prognostic factors in glioblastomamultiforme. 10 years experience of a single institution. Strah-lenther Onkol 177:283–290

Kaaijk P, Troost D, Das PK, Leenstra S, Bosch DA (1995) Longterm culture of organotypic multicellular glioma spheroids: agood alternative culture model for studying gliomas. Neuro-pathol Appl Neurobiol 21:386–391

Kaaijk P, Troost D, Sminia P, Hulshof MC, van der Kracht AH,Leenstra S, Bosch DA (1997) Hypofractionated radiation in-duces a decrease in cell proliferation but no histological damageto organotypic multicellular spheroids of human glioblastomas.Eur J Cancer 33:645–651

Kerr JFK, Wyllie AH, Currie AH (1972) Apoptosis, a basic bio-logical phenomenon with wider implications in tissue kinetics.Br J Cancer 26:239–245

Kleynen CE, Stoter TR, Tadema TM, Stalpers LJ, Dirven CMF,Leenstra, S, Van der Valk P, Slotman BJ, Sminia P(2003) The effects of irradiation on cell-migration from glio-blastoma multiforme biopsy spheroids. Anticancer Res23:4909–4912

Kunz-Schughart LA, Kreutz M, Knuechel R (1998) Multicellularspheroids: a three-dimensional in vitro culture system to studytumour biology. Int J Exp Path 79:1–23

Mahesparan R, Tysnes BB, Read TA, Enger PO, Bjerkvig R,Lund-Johansen M (1999) Extracellular matrix-induced cellmigration from glioblastoma biopsy specimens in vitro. ActaNeuropathol 97:231–239

Mueller-Klieser WF, Freyer JP, Sutherland RM (1986) Influence ofglucose and oxygen supply conditions on the oxygenation ofmulticellular spheroids. Br J Cancer 53:345–353

O‘Connor PM, Fan S (1996) Cell cycle checkpoints and cancerchemotherapy. In: Pinedo HM, Schornagel JH (eds) Platinumand other metal coordination compounds in cancer chemo-therapy 2. Plenium Press, New York, pp 293–301

Olive PL, Durand RE (1994) Drug and radiation resistance inspheroids: cell contact and kinetics. Cancer Metastasis Rev13:121–138

Ono Y, Tamiya T, Ichikawa T, Matsumoto K, Furuta T, OhmotoT, Akiyama K, Seki S, Ueki K, Louis DN (1997) Accumulationof wild type p53 in astrocytomas is associated with increasedp21 expression. Acta Neuropathol 94:21–27

Reichert M, Steinbach JP, Supra P, Weller M (2002) Modulation ofgrowth and radiochemosensitivity of human malignant gliomacells by acidosis. Cancer 95:1113–1119

Santini MT, Rainaldi G, Indovina PL (1999) Multicellular tumourspheroids in radiation biology. Int J Radiat Biol 75:787–799

Sheleg SV, Korotkevich EA, Zhavrid EA, Muravskaya GV,Smeyanovich AF, Shanko YG, Yurkshtovich TL, BychkovskyPB, Belyaev SA (2002) Local chemotherapy with cisplatin-de-pot for glioblastoma multiforme. J Neurooncol 60:53–59

Sminia P, Acker H, Eikesdal, HP, Kaaijk P, Enger PO, Slotman,BJ, Bjerkvig R (2003) Oxygenation and response to irradiation

731

of organotypic multicellular spheroids of human glioma.Anticancer Res 23:1461–1466

Stuschke M, Budach V, Sack H (1993) Radioresponsiveness ofhuman glioma, sarcoma and breast cancer spheroids dependson tumor differentiation. Int J Radiat Oncol Biol Phys 27:627–636

Sutherland RM (1986) Importance of critical metabolism and cel-lular interactions in the biology of microregions of tumors.Cancer 58:1668–1680

Sutherland RM (1988) Cell and environment interactions in tumormicroregions: the multicell spheroid model. Science 240:177–184

Taghian A, Ramsay J, Allalunis-Turner J, Budach W, Gioioso D,Pardo F, Okunieff P, Bleehen N, Urtasun R, Suit H (1993)Intrinsic radiation sensitivity may not be the major determinantof the poor clinical outcome of glioblastoma multiforme. Int JRadiat Oncol Biol Phys 25:243–249

Taghian A, DuBois W, Budach W, Baumann M, Freeman J, SuitH (1995) In vivo radiation sensitivity of glioblastoma multi-forme. Int J Radiat Oncol Biol Phys 32:99–104

Walenta S, Doetsch J, Mueller-Klieser W, Kunz-Schughart LA(2000) Metabolic imaging in multicellular spheroids of onco-gene-transfected fibroblasts. J Histochem Cytochem 48:509–522

Wilkins DE, Ng CH, Raaphorst GP (1996) Cisplatin and low doserate iradiation in cisplatin resistant and sensitive human gliomacells. Int J Radiat Oncol Biol Phys 36:105–111

Wolff JE, Galla HJ, Poppenborg H (1998) Long recovery timesimprove the detection of cellular resistance in vitro. J CancerRes Clin Oncol 124:409–414

Wolff JE, Trilling T, Molenkamp G, Egeler RM, Jurgens H (1999)Chemosensitivity of glioma cells in vitro: a meta analysis. JCancer Res Clin Oncol 125:481–486

Yount GL, Haas-Kogan DA, Levine KS, Aldape KD, Israel MA(1998) Ionizing radiation inhibits chemotherapy-inducedapoptosis in cutured glioma cells: implications for combinedmodality therapy. Cancer Res 58:3819–3825

Zatterstrom UK, Engellau J, Johansson MC, Wennerberg J,Kjellen E (1995) Radiation effects on S-phase duration, label-ling index, potential doubling time and DNA distribution inhead and neck cancer xenografts. Acta Oncol 34:205–211

732