+ All Categories
Home > Documents > Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

Date post: 23-Dec-2016
Category:
Upload: chitra
View: 215 times
Download: 1 times
Share this document with a friend
8
CLINICAL STUDY Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas Rakesh Kumar Gupta Mehar C. Sharma Vaishali Suri Aanchal Kakkar Manmohan Singh Chitra Sarkar Received: 9 July 2013 / Accepted: 21 October 2013 / Published online: 1 November 2013 Ó Springer Science+Business Media New York 2013 Abstract Ependymomas are relatively uncommon tumours of the central nervous system which arise from the ependymal lining of the ventricles and spinal canal. The molecular changes leading to ependymal oncogenesis are not completely understood. We examined chromosome 9q33-34 locus for gain, potential oncogenes at this locus (Notch-1 and Tenascin-C) and Notch pathway target genes (Hes-1, Hey-2 & C-myc) in ependymomas by fluorescent in situ hybridization (FISH) and immunohistochemistry (IHC), respectively, to assess if they have any correlation with clinical characteristics. We analyzed 50 cases of ependymomas by FISH for 9q gain and by IHC for Notch-1 and its target gene proteins (Hes-1, Hey-2 and C-myc) expression. We also performed IHC for Tenascin-C to rule out any correlation with aggressiveness/grade of tumour. FISH study revealed significant chromosome 9q gain in ependymomas of adult onset (age [ 18 years) and spinal cord origin. Notch-1 showed significantly more frequent immunohistochemical expression in supratentorial and anaplastic ependymomas. Tenascin-C (TN-C) expression was significant in intracranial, childhood (age B 18 years) and anaplastic ependymomas. Of the three Notch pathway target gene proteins (Hes-1, Hey-2 and C-myc), Hes-1 and C-myc expression showed significant correlation with ana- plastic and adult onset ependymomas, respectively. Genetic alterations are independent prognostic markers in ependymomas. A clinicopathological correlation with vari- ous molecular signatures may be helpful in the development of new therapeutic targets. Keywords Ependymoma Á FISH Á IHC Á Chromosome 9q Á Notch-1 Introduction Ependymomas are glial tumours which originate from the ependymal lining of the ventricles and central canal of the spinal cord [1]. They are the third most common paediatric central nervous system tumours accounting for 6–12 % of all intracranial tumours in children [24]. The prognosis of childhood ependymomas is relatively poor, with 5-year overall survival (OS) and progression-free survival (PFS) rates ranging between 50–60 and 30–50 %, respectively [2, 4]. A large meta-analysis on 2,408 ependymoma patients identified WHO grade as an independent prognostic factor [5], whereas some studies suggested grading is a matter of subjectivity [4, 69]. More than 50 % of cases occur in children under 5 years of age, which is a potential hurdle for the use of radiotherapy because of its long term complica- tions, and the beneficial role of chemotherapy is controver- sial [2, 6, 1013]. Patient outcome prediction is difficult because of heterogeneity in clinical behavior and molecular changes, and the results of existing studies on prognostic markers are contradictory [14, 15]. Therefore, there is need to improve the understanding of the biology of ependymo- mas in order to develop new therapeutic targets and agents. Previous studies have demonstrated the role of Notch pathway in the pathogenesis of central nervous system tumours including glioblastoma multiforme and medullo- blastoma, and in several non-neural tumours [1619]. R. K. Gupta Á M. C. Sharma (&) Á V. Suri Á A. Kakkar Á C. Sarkar Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India e-mail: [email protected] M. Singh Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi 110029, India 123 J Neurooncol (2014) 116:267–274 DOI 10.1007/s11060-013-1287-z
Transcript
Page 1: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

CLINICAL STUDY

Study of chromosome 9q gain, Notch pathway regulatorsand Tenascin-C in ependymomas

Rakesh Kumar Gupta • Mehar C. Sharma •

Vaishali Suri • Aanchal Kakkar • Manmohan Singh •

Chitra Sarkar

Received: 9 July 2013 / Accepted: 21 October 2013 / Published online: 1 November 2013

� Springer Science+Business Media New York 2013

Abstract Ependymomas are relatively uncommon

tumours of the central nervous system which arise from the

ependymal lining of the ventricles and spinal canal. The

molecular changes leading to ependymal oncogenesis are

not completely understood. We examined chromosome

9q33-34 locus for gain, potential oncogenes at this locus

(Notch-1 and Tenascin-C) and Notch pathway target genes

(Hes-1, Hey-2 & C-myc) in ependymomas by fluorescent

in situ hybridization (FISH) and immunohistochemistry

(IHC), respectively, to assess if they have any correlation

with clinical characteristics. We analyzed 50 cases of

ependymomas by FISH for 9q gain and by IHC for Notch-1

and its target gene proteins (Hes-1, Hey-2 and C-myc)

expression. We also performed IHC for Tenascin-C to rule

out any correlation with aggressiveness/grade of tumour.

FISH study revealed significant chromosome 9q gain in

ependymomas of adult onset (age [ 18 years) and spinal

cord origin. Notch-1 showed significantly more frequent

immunohistochemical expression in supratentorial and

anaplastic ependymomas. Tenascin-C (TN-C) expression

was significant in intracranial, childhood (age B 18 years)

and anaplastic ependymomas. Of the three Notch pathway

target gene proteins (Hes-1, Hey-2 and C-myc), Hes-1 and

C-myc expression showed significant correlation with ana-

plastic and adult onset ependymomas, respectively. Genetic

alterations are independent prognostic markers in

ependymomas. A clinicopathological correlation with vari-

ous molecular signatures may be helpful in the development

of new therapeutic targets.

Keywords Ependymoma � FISH � IHC �Chromosome 9q � Notch-1

Introduction

Ependymomas are glial tumours which originate from the

ependymal lining of the ventricles and central canal of the

spinal cord [1]. They are the third most common paediatric

central nervous system tumours accounting for 6–12 % of all

intracranial tumours in children [2–4]. The prognosis of

childhood ependymomas is relatively poor, with 5-year

overall survival (OS) and progression-free survival (PFS)

rates ranging between 50–60 and 30–50 %, respectively

[2, 4]. A large meta-analysis on 2,408 ependymoma patients

identified WHO grade as an independent prognostic factor

[5], whereas some studies suggested grading is a matter of

subjectivity [4, 6–9]. More than 50 % of cases occur in

children under 5 years of age, which is a potential hurdle for

the use of radiotherapy because of its long term complica-

tions, and the beneficial role of chemotherapy is controver-

sial [2, 6, 10–13]. Patient outcome prediction is difficult

because of heterogeneity in clinical behavior and molecular

changes, and the results of existing studies on prognostic

markers are contradictory [14, 15]. Therefore, there is need

to improve the understanding of the biology of ependymo-

mas in order to develop new therapeutic targets and agents.

Previous studies have demonstrated the role of Notch

pathway in the pathogenesis of central nervous system

tumours including glioblastoma multiforme and medullo-

blastoma, and in several non-neural tumours [16–19].

R. K. Gupta � M. C. Sharma (&) � V. Suri � A. Kakkar �C. Sarkar

Department of Pathology, All India Institute of Medical

Sciences, New Delhi 110029, India

e-mail: [email protected]

M. Singh

Department of Neurosurgery, All India Institute of Medical

Sciences, New Delhi 110029, India

123

J Neurooncol (2014) 116:267–274

DOI 10.1007/s11060-013-1287-z

Page 2: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

However, not much literature is available on the involve-

ment of this pathway in ependymomas. Therefore, we

undertook this study to analyze the expression of Notch

and its target genes in ependymomas.

Materials and methods

We retrieved 50 paraffin embedded tissue blocks of cases

diagnosed as ependymoma in the Department of Pathology

during a period of 10 years (2002–2011). Diagnoses were

confirmed by histopathological assessment of H & E

stained slides by three neuropathologists (RKG, MCS, CS),

according to the 2007 WHO classification of CNS tumours

[1]. Cases with sufficient material in tissue blocks and

diagnostic concurrence were included in the study.

Tumor tissue had been received in neutral buffered

formalin and was routinely processed and paraffin

embedded. Five micron thick sections were cut for hema-

toxylin & eosin (H&E) staining and for immunohisto-

chemistry. Immunohistochemical studies were performed

using antibodies directed against Notch-1(dilution, 1:200,

AntiNotch-1, EP1238Y, Abcam, USA), Tenascin-C (dilu-

tion, 1:200, Anti Tenascin-C, MAB19101, Millipore,

USA), Hes-1(dilution, 1:200, Anti Hes-1, ab71559, Ab-

cam, USA), Hey-2 (dilution, 1:25, Anti Hey-2,

HPA030205, Sigma, USA) and C-myc (dilution, 1:100,

Anti C-myc, 9E10, Santa Cruz, USA). Labeled streptavidin

biotin kit Universal (Dako, Denmark) was used as a

detection system. Antigen retrieval was performed in a

microwave oven using citrate buffer at pH 6.0 for all

antibodies except Tenascin-C, for which Trypsin digestion

(TA-125-TR, Thermo Fisher Scientific, UK) was done. For

each batch, appropriate positive and negative controls were

taken. Immunohistochemistry with all antibodies was

interpreted as follows: 0 % - grade 0; 1–25 % - grade 1;

26–50 % - grade 2; [50 % - grade 3.

Fluorescent in situ hybridization analysis

FISH study was performed on 50 paraffin embedded tissue

samples. Chromosome 9q was studied using a centromeric

probe (CEP9) and dual colour, dual fusion bcr/abl trans-

location probe for 9q33-34 locus (M/S Vysis, Inc., Abbott

Laboratories SA, Downers Grove, IL, USA). Sections were

deparaffinized by immersing in xylene thrice for 10-min

each, followed by two 3-min immersions in 100 % ethanol.

Following rinsing in water, target retrieval was achieved

using citrate buffer, pH 6.0 and boiling in a microwave for

20 min. Slides were exposed to 0.04 % pepsin (P-7000;

Sigma- Aldrich, St. Louis, MO) digestion for 30 min at

37 �C, fixed and dehydrated. Probe mixture (10 ll per

slide) was applied on each section. Simultaneous probe/

specimen denaturation at 73 �C for 5 min with subsequent

overnight incubation at 37 �C was performed in Ther-

mobrite TM hybridization chamber (Vysis Inc). The sec-

tions were washed the next day in 2X SSC (2 min at 73 �C)

followed by 0.5X SSC (2 min at room temperature) and

counterstained with 4,6-diamidino-2-phenylindole (Vysis,

Inc) and visualized under a fluorescent microscope. Signals

were scored at least in 200 non-overlapping, intact nuclei

and the number of test (orange) and control (aqua) signals

was noted. Nuclei with two test and two control signals

were regarded as normal. Cases with [10 % nuclei hav-

ing [3 test signals in comparison to the control signals

were considered as demonstrating gain of 9q33-34. This

cut-off value of 10 % was decided based on study in nor-

mal brain samples from intractable epilepsy cases.

Statistical analysis

Relationships among variables were assessed using stan-

dard statistical techniques: Fisher’s exact test and Chi

square test. We considered a relationship significant at a

p value less than 0.05.

Results

Clinicopathologic parameters (Table 1)

The 50 tumour samples were comprised of 44 % (22/50)

children and 56 % (28/50) adults. Male to female ratio

was 2:1. In the pediatric group (B18 years), the age

ranged from 2 to 18 years with mean age of 9.1 years,

while in the adult group ([18 years) the age ranged from

19 to 62 years with mean age of 35.9 years. Out of 50

cases, 12 % (6 cases), 52 % (26 cases) and 36 % (18

cases) were of grade I, grade II and grade III, respec-

tively. Site distribution of the cases were 30 % (15 cases)

supratentorial, 32 % (16 cases) infratentorial and 38 %

(19 cases) intraspinal.

Chromosomal 9q33-34 gain in ependymomas

Chromosome 9q gain (Fig. 1) was identified in 48 %

(24/50) of patients, of which 37.5 % (9/24) were intracra-

nial and 62.5 % (15/24) were intraspinal (Fig. 2). This

difference was statistically significant (p = 0.005). Of the

24 patients who showed 9q33-34 gain, 29.1 % (7 cases)

were children and 70.8 % (17 cases) were adults, and this

difference was also statistically significant (p = 0.03).

Although correlation between chromosome 9q gain and

tumour grade was not statistically significant, it showed a

decreasing trend with increase in tumour grade.

268 J Neurooncol (2014) 116:267–274

123

Page 3: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

Table 1 Clinicopathological features of the cases with their FISH status

Sample Age (years) Sex Site Surgery Grade (WHO) OS (months) Status FISH 9q gain

1 46 F IS GTR I 30 – Yes

5 41 M IS SR I 4 – Yes

30 55 F IS SR I 22.2 ADF Yes

38 21 M IS SR I 12 ADF Yes

49 9 M IS GTR I – – No

50 17 M ST SR I 6 ADF No

4 60 F PF GTR II 2 DOD Yes

7 4 M PF SR II – – No

8 19 M PF SR II 8.2 – No

10 33 F PF SR II 21.2 ADF No

11 35 M IS GTR II 33.5 ADF Yes

12 52 M IS SR II 27.3 ADF Yes

13 2 M PF GTR II 3 – No

15 44 M IS GTR II – – No

16 45 F IS GTR II 15.8 ADF No

17 22 M IS GTR II 24 ADF No

20 5 M PF SR II 3.5 – No

21 9 M PF SR II 35 ADF No

22 23 M IS GTR II – – No

23 18 M PF GTR II 4 – Yes

24 10 M ST GTR II 9.4 ADF Yes

25 25 F IS GTR II 14 ADF Yes

28 35 M IS GTR II – – Yes

31 12 M PF GTR II 3 ADF Yes

32 33 F IS GTR II 2.7 ADF Yes

34 22 M IS GTR II – – Yes

35 30 M IS GTR II 15 ADF Yes

37 30 M IS GTR II 16.5 ADF Yes

39 34 M IS GTR II – – Yes

42 9 M PF GTR II 22.5 ADF No

44 7 F ST SR II 10.6 ADF No

46 13 M ST SR II 20.8 ADF No

2 60 M ST GTR III – – No

3 35 M PF GTR III – – Yes

6 22 F ST GTR III – – No

9 30 M ST GTR III 23.6 ADF No

14 8 F ST GTR III – – No

18 28 M ST SR III – – No

19 1 F PF GTR III 15.5 ADF No

26 10 F ST GTR III 13.3 ADF Yes

27 10 F IS GTR III 23.4 ADF Yes

29 4 M PF SR III 22.4 ADF Yes

33 62 F PF GTR III 17.3 ADF Yes

36 12 M ST GTR III 18 ADF Yes

40 2 M PF SR III 2 DOD No

41 24 M PF SR III 8.5 – No

43 40 M ST SR III 13.4 ADF No

J Neurooncol (2014) 116:267–274 269

123

Page 4: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

Immunohistochemical analysis of candidate oncogenes

and target genes (Table 2; Fig. 3)

Notch-1 showed significantly higher immunoexpression in

supratentorial tumours in comparison to infratentorial

(p = 0.001) and intraspinal (p = 0.01) tumours. However,

this difference was not statistically significant between

intracranial (supratentorial & infratentorial) and intraspinal

tumours. Immunoexpression of TN-C was significantly

higher in intracranial tumours in comparison to intraspinal

tumours (p = 0.05). Notch-1, TN-C and Hes-1 showed a

significantly higher expression in grade III tumours in

comparison to grade II tumours with a p value of 0.001,

0.006 and 0.02, respectively. Notch-1 showed increased

expression with tumour grade and the difference between

grade I & grade III tumours was statistically significant

(p = 0.004). TN-C immunoexpression was seen more

commonly in childhood tumours in comparison to adults

(p = 0.003). C-myc immunoexpression was contrary to

Fig. 1 Representative fluorescent in situ hybridization (FISH) show-

ing gain of 9q33-34 (red signals) as compared to control blue signals

(CEP9). Green signals represent chromosome 22 ‘BCR’ region and

are not part of this study

Fig. 2 Flowchart showing

clinicopathological correlation

of the cases with 9q33-34 gain

Table 1 continued

Sample Age (years) Sex Site Surgery Grade (WHO) OS (months) Status FISH 9q gain

45 9 F ST GTR III 17.5 ADF No

47 3 F ST GTR III 17 ADF No

48 12 M ST GTR III – – No

M male, F female, IS intraspinal, PF posterior fossa, ST supratentorial, GTR gross total resection, SR subtotal resection, ADF alive disease free,

DOD died of disease

270 J Neurooncol (2014) 116:267–274

123

Page 5: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

TN-C expression and more commonly seen in tumours

which occurred in adults as compared to childhood

tumours (p = 0.03). None of the oncogenes (Notch-1 and

TN-C) and target genes (Hes-1, Hey-2 & C-myc) showed

correlation with chromosome 9q gain.

Discussion

Several cytogenetic and molecular studies have been pub-

lished, highlighting the chromosomal and molecular alter-

ations in ependymomas, but have failed to show specific

target genes involved in ependymal oncogenesis, unlike

astrocytic gliomas. This failure of demonstrating specific

molecular changes proved to be a hindrance in developing

specific targeted therapies in ependymomas. These tumours

frequently occur in young children, and the usual chemo-

therapeutic agents and radiotherapy have high toxicity,

resulting in long term complications. Therefore, this

necessitates the urgent unraveling of molecular alterations

in ependymomas to discover newer therapeutic agents.

In the last decade, several authors have studied cyto-

genetic changes in ependymomas either by conventional

comparative genomic hybridization (CGH) on metaphase

chromosomes, karyotyping or array based expression pro-

filing CGH [20–32]. A few studies have shown gain of 9q

in ependymomas [20–24, 26]. Reardon et al. [20] studied

32 cases of ependymomas by CGH and found either gain of

chromosome 1q or 9. Hirose et al. [21] found that gains of

1q and loss of 9 were preferentially associated with grade III

ependymomas. None of their cases showed 9q gain. Jeuken

et al. [24] recorded 9q gain more commonly in posterior

fossa ependymomas and in adult patients. Rousseau et al.

[31] have shown that combination of chromosome 9q gain

in association with other chromosomal gains and losses is

more frequently seen in spinal cord grade II and myxo-

papillary grade I ependymomas. However, the exact sig-

nificance of this gain was not demonstrated in these studies.

To date, only a single study has examined 9q gain along

with expression of oncogenes identified at this locus, and

their downstream targets. Puget et al. performed bacterial

artificial chromosome (BAC) based array-comparative

genomic hybridization(aCGH) on 59 paediatric ependy-

moma samples and found that gain of chromosome 9q is

frequently associated with relapse, age older than 3 years

and posterior fossa location. This gain was further localized

at 9q33-34 region by FISH. Analysis of various genes at

this locus confirmed mutations of TN-C and Notch-1 genes

located in this region [28]. These authors have shown that

Notch pathway activation leads to overexpression of target

genes Hes-1, Hey-2 and C-myc which conferred a growth

advantage on the tumour cells. In our study, we identified

chromosome 9q gain by FISH in a significant proportion of

ependymomas (48 %). We also found a significantly higher

percentage of chromosome 9q33-34 gain in spinal cord

ependymomas, including myxopapillary. Our results are

similar to previous studies, [21, 22, 31, 33] but contrary to

that of Puget et al. [30]. In the present series, chromosome

9q gain was observed more commonly in adult patients

(p = 0.03), as shown in earlier studies [20, 22, 34]. There

was no significant difference in frequency of 9q gain in

different tumour grades, however there was a decreasing

trend in 9q gain with increase in tumour grade, and these

results are in concordance with published studies [32, 35].

We identified significant upregulation of Notch-1

expression in supratentorial tumours, similar to previous

studies [28–30, 36]. Modena et al. [28] in 2006, showed

overexpression of Notch pathway proteins in supratentorial

ependymomas. TN-C, which is involved in central nervous

system embryogenesis, expression was significantly more

frequent in ependymomas which were located intracrani-

ally, as well as in childhood tumours, and these results

were in concordance to earlier studies [28, 30]. Overex-

pression of TN-C has been shown in many glial tumours,

and this expression has been shown to be associated with

increased vascularity, poor prognosis and relapse [37, 38].

In the present series, higher expression of TN-C, Notch-1,

as well as its target gene Hes-1 was observed in anaplastic

ependymomas (grade III) in comparison to grade I and

grade II. These results were similar to the observation

made by Puget et al. [30]. The discordance between

chromosome 9q gain and Notch-1 & TN-C oncogenes

overexpression at different sites may be due to activating or

inactivating mutations in these genes [30].

Table 2 Correlation of various gene proteins expression with age, sex and grade of the tumour

Markers No. of positive

cases (n = 50)

Age Site Grade

Children Adults ST PF IS I II III

TNC 16 (32 %) 12 (75.0 %) 4 (25.0 %) 5 (31.2 %) 8 (50.0 %) 3 (18.8 %) 3 (18.8 %) 5 (31.2 %) 8 (50.0 %)

Notch1 18 (36.0 %) 7 (38.8 %) 11 (61.1 %) 11 (61.1 %) 2 (11.1 %) 5 (27.7 %) 1 (5.5 %) 5 (27.7 %) 12 (66.6 %)

HES1 20 (40.0 %) 7 (35.0 %) 13 (65.0 %) 3 (15.0 %) 7 (35.0 %) 10 (50.0 %) 3 (15.0 %) 14 (70.0 %) 3 (15.0 %)

HEY2 31 (62.0 %) 12 (38.7 %) 19 (61.2 %) 10 (32.2 %) 10 (32.2 %) 11 (35.4 %) 2 (6.4 %) 16 (51.6 %) 13 (41.9 %)

c-myc 26 (52.0 %) 5 (19.2 %) 21 (80.7 %) 7 (26.9 %) 7 (26.9 %) 12 (46.1 %) 4 (15.3 %) 14 (53.8 %) 8 (30.6 %)

J Neurooncol (2014) 116:267–274 271

123

Page 6: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

272 J Neurooncol (2014) 116:267–274

123

Page 7: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

To summarize, we found a significantly higher expres-

sion of Notch-1 and TN-C in supratentorial and intracranial

(both supratentorial & posterior fossa) tumours, respec-

tively, as well as in anaplastic ependymomas. These

markers can help in risk stratification of ependymomas and

may further serve as targets for novel therapeutic agents.

Immunotherapy using radiolabelled anti TN-C antibodies

has shown promising results in hematological malignancies

and brain tumours [39, 40]. c-Secretase (a Notch pathway

enzyme) inhibitors might be an important therapeutic

option in future for supratentorial ependymomas [30].

Ependymomas can be categorized based on their unique

molecular profiles at different site, grade and age groups;

accordingly target oriented therapeutic interventions might

be developed in near future.

Acknowledgments We would like to acknowledge Mr Sujit for

help in performing FISH studies, Mr Pankaj & Mrs Kiran for helping

in immmunohistochemistry. Our special thanks to Mr Guresh for

statistical analysis.

Conflict of interest None.

References

1. McLendon RE, Wiestler OD, Kros JM, Korshunov A, Ng HK

(2007) Anaplastic ependymoma. In: Louis DN, Ohgaki H,

Wiestler OD, Cavenee WK (eds) WHO classification of tumours

of the central nervous system, vol 4. IARC Press, Lyon, pp 79–80

2. Bouffet E, Perilongo G, Canete A, Massimino M (1998) Intra-

cranial ependymomas in children: a critical review of prognostic

factors and a plea for cooperation. Med Pediatr Oncol 30:

319–331

3. Miller RW, Young JL Jr, Novakovic B (1995) Childhood cancer.

Cancer 75:395–405

4. Merchant TE (2002) Current management of childhood ependy-

moma. Oncology (Williston Park) 16:629–648

5. Rodrıguez D, Cheung MC, Housri N, Quinones-Hinojosa A,

Camphausen K, Koniaris LG (2009) Outcomes of malignant CNS

ependymomas: an examination of 2408 cases through the Sur-

veillance, Epidemiology, and End Results (SEER) database

(1973–2005). J Surg Res 156:340–351

6. Messahel B, Ashley S, Saran F, Ellison D, Ironside J, Phipps K,

Cox T, Chong WK, Robinson K, Picton S, Pinkerton CR, Mal-

lucci C, Macarthur D, Jaspan T, Michalski A, Grundy RG,

Children’s Cancer Leukaemia Group Brain Tumour Committee

(2009) Relapsed intracranial ependymoma in children in the UK:

patterns of relapse, survival and therapeutic outcome. Eur J

Cancer 45:1815–1823

7. Figarella-Branger D, Civatte M, Bouvier-Labit C, Gouvernet J,

Gambarelli D, Gentet JC, Lena G, Choux M, Pellissier JF (2000)

Prognostic factors in intracranial ependymomas in children.

J Neurosurg 93:605–613

8. Korshunov A, Golanov A, Sycheva R, Timirgaz V (2004) The

histologic grade is a main prognostic factor for patients with

intracranial ependymomas treated in the microneurosurgical era:

an analysis of 258 patients. Cancer 100:1230–1237

9. Kurt E, Zheng PP, Hop WC, van der Weiden M, Bol M, van den

Bent MJ, Avezaat CJ, Kros JM (2006) Identification of relevant

prognostic histopathologic features in 69 intracranial ependy-

momas, excluding myxopapillary ependymomas and subepend-

ymomas. Cancer 106:388–395

10. Duffner PK, Krischer JP, Sanford RA, Horowitz ME, Burger PC,

Cohen ME, Friedman HS, Kun LE (1998) Prognostic factors in

infants and very young children with intracranial ependymomas.

Pediatr Neurosurg 28:215–222

11. Grill J, Le Deley MC, Gambarelli D, Raquin MA, Couanet D,

Pierre-Kahn A, Habrand JL, Doz F, Frappaz D, Gentet JC, Edan

C, Chastagner P, Kalifa C, French Society of Pediatric Oncology

(2000) Postoperative chemotherapy without irradiation for

ependymoma in children under 5 years of age: a multicenter trial

of the French Society of Pediatric Oncology. J Clin Oncol

19:1288–1296

12. Heidemann RLPR, Albright LA, Freeman CR, Rorke LB (1997)

Tumors of the central nervous system, 1st edn. Lippincott-Raven,

Philadelphia

13. Grundy RG, Wilne SA, Weston CL, Robinson K, Lashford LS,

Ironside J, Cox T, Chong WK, Campbell RH, Bailey CC, Gatt-

amaneni R, Picton S, Thorpe N, Mallucci C, English MW, Punt

JA, Walker DA, Ellison DW, Machin D, Children’s Cancer and

Leukaemia Group (formerly UKCCSG) Brain Tumour Commit-

tee (2007) Primary postoperative chemotherapy without radio-

therapy for intracranial ependymoma in children: the UKCCSG/

SIOP prospective study. Lancet Oncol 8:696–705

14. Hamilton RL, Pollack IF (1997) The molecular biology of

ependymomas. Brain Pathol 7:807–822

15. Tabori U, Vukovic B, Zielenska M, Hawkins C, Braude I, Rutka

J, Bouffet E, Squire J, Malkin D (2006) The role of telomere

maintenance in the spontaneous growth arrest of pediatric low-

grade gliomas. Neoplasia 8:136–142

16. Stockhausen MT, Kristoffersen K, Poulsen HS (2010) The

functional role of Notch signaling in human gliomas. Neuro

Oncol 12(2):199–211

17. Ying M, Wang S, Sang Y, Sun P, Lal B, Goodwin CR, Guerrero-

Cazares H, Quinones-Hinojosa A, Laterra J, Xia S (2011) Reg-

ulation of glioblastoma stem cells by retinoic acid: role for Notch

pathway inhibition. Oncogene 30(31):3454–3467

18. Fan X, Mikolaenko I, Elhassan I, Ni X, Wang Y, Ball D, Brat DJ,

Perry A, Eberhart CG (2004) Notch1 and notch2 have opposite

effects on embryonal brain tumor growth. Cancer Res 64(21):

7787–7793

19. Hallahan AR, Pritchard JI, Hansen S, Benson M, Stoeck J, Hatton

BA, Russell TL, Ellenbogen RG, Bernstein ID, Beachy PA, Ol-

son JM (2004) The SmoA1 mouse model reveals that notch

signaling is critical for the growth and survival of sonic hedge-

hog-induced medulloblastomas. Cancer Res 64(21):7794–7800

20. Reardon DA, Entrekin RE, Sublett J, Ragsdale S, Li H, Boyett J,

Kepner JL, Look AT (1999) Chromosome arm 6q loss is the most

common recurrent autosomal alteration detected in primary

pediatric ependymoma. Genes Chromosomes Cancer 24:230–237

21. Hirose Y, Aldape K, Bollen A et al (2001) Chromosomal

abnormalities subdivide ependymal tumors into clinically rele-

vant groups. Am J Pathol 158:1137–1143

22. Carter M, Nicholson J, Ross F, Crolla J, Allibone R, Balaji V,

Perry R, Walker D, Gilbertson R, Ellison DW (2002) Genetic

abnormalities detected in ependymomas by comparative genomic

hybridisation. Br J Cancer 18(86):929–939

Fig. 3 Immunohistochemistry showing expression of various pro-

teins in grade I myxopapillary ependymoma (a H&E, b TNC,

c Notch1, d HES1, e HEY2, f c-myc), grade II ependymoma (g H&E,

h TNC, i Notch1, j HES1, k HEY2, l c-myc), and grade III

ependymoma (m H&E, n TNC, o Notch1, p HES1, q HEY2, r c-

myc), (9400 each)

b

J Neurooncol (2014) 116:267–274 273

123

Page 8: Study of chromosome 9q gain, Notch pathway regulators and Tenascin-C in ependymomas

23. Dyer S, Prebble E, Davison V, Davies P, Ramani P, Ellison D,

Grundy R (2002) Genomic imbalances in pediatric intracranial

ependymomas define clinically relevant groups. Am J Pathol

161:2133–2141

24. Jeuken JW, Sprenger SH, Gilhuis J, Teepen HL, Grotenhuis AJ,

Wesseling P (2002) Correlation between localization, age, and

chromosomal imbalances in ependymal tumours as detected by

CGH. J Pathol 197:238–244

25. Mendrzyk F, Korshunov A, Benner A, Toedt G, Pfister S, Rad-

lwimmer B, Lichter P (2006) Identification of gains on 1q and

epidermal growth factor receptor overexpression as independent

prognostic markers in intracranial ependymoma. Clin Cancer Res

12:2070–2079

26. Pezzolo A, Capra V, Raso A, Morandi F, Parodi F, Gambini C,

Nozza P, Giangaspero F, Cama A, Pistoia V, Garre ML (2008)

Identification of novel chromosomal abnormalities and prognos-

tic cytogenetics markers in intracranial pediatric ependymoma.

Cancer Lett 18(261):235–243

27. Ammerlaan AC, de Bustos C, Ararou A, Buckley PG, Mantri-

pragada KK, Verstegen MJ, Hulsebos TJ, Dumanski JP (2005)

Localization of a putative low-penetrance ependymoma suscep-

tibility locus to 22q11 using a chromosome 22 tiling-path geno-

mic microarray. Genes Chromosomes Cancer 43:329–338

28. Modena P, Lualdi E, Facchinetti F et al (2006) Identification of

tumor-specific molecular signatures in intracranial ependymoma

and association with clinical characteristics. J Clin Oncol

24:5223–5233

29. Palm T, Figarella-Branger D, Chapon F, Lacroix C, Gray F, Sca-

ravilli F, Ellison DW, Salmon I, Vikkula M, Godfraind C (2009)

Expression profiling of ependymomas unravels localization and

tumor grade-specific tumorigenesis. Cancer 115:3955–3968

30. Puget S, Grill J, Valent A, Bieche I, Dantas-Barbosa C, Kauff-

mann A, Dessen P, Lacroix L, Geoerger B, Job B, Dirven C,

Varlet P, Peyre M, Dirks PB, Sainte-Rose C, Vassal G (2009)

Candidate genes on chromosome 9q33-34 involved in the pro-

gression of childhood ependymomas. J Clin Oncol 27:1884–1892

31. Rousseau A, Idbaih A, Ducray F, Criniere E, Fevre-Montange M,

Jouvet A, Delattre JY (2010) Specific chromosomal imbalances as

detected by array CGH in ependymomas in association with tumor

location, histological subtype and grade. J Neurooncol 97:353–364

32. Korshunov A, Witt H, Hielscher T, Benner A, Remke M, Ry-

zhova M, Milde T, Bender S, Wittmann A, Schottler A, Kulozik

AE, Witt O, von Deimling A, Lichter P, Pfister S (2010)

Molecular staging of intracranial ependymoma in children and

adults. J Clin Oncol 28:3182–3190

33. Mahler-Araujo MB, Sanoudou D, Tingby O, Liu L, Coleman N,

Ichimura K, Collins VP (2003) Structural genomic abnormalities

of chromosomes 9 and 18 in myxopapillary ependymomas.

J Neuropathol Exp Neurol 62:927–935

34. Kilday JP, Rahman R, Dyer S, Ridley L, Lowe J, Coyle B,

Grundy R (2009) Pediatric ependymoma: biological perspectives.

Mol Cancer Res 7:765–786

35. Scheil S, Bruderlein S, Eicker M, Herms J, Herold-Mende C,

Steiner HH, Barth TF, Moller P (2001) Low frequency of chro-

mosomal imbalances in anaplastic ependymomas as detected by

comparative genomic hybridization. Brain Pathol 11:133–143

36. Taylor MD, Poppleton H, Fuller C et al (2005) Radial glia cells

are candidate stem cells of ependymoma. Cancer Cell 8:323–335

37. Herold-Mende C, Mueller MM, Bonsanto MM, Schmitt HP,

Kunze S, Steiner HH (2000) Clinical impact and functional

aspects of tenascin-C expression during glioma progression. Int J

Cancer 20(98):362–369

38. Korshunov A, Golanov A, Timirgaz V (2000) Immunohisto-

chemical markers for prognosis of ependymal neoplasms. J Neu-

rooncol 58:255–270

39. Huntly BJ, Gilliland DG (2005) Leukaemia stem cells and the

evolution of cancer-stem-cell research. Nat Rev Cancer 5:311–321

40. Reardon DA, Akabani G, Coleman RE et al (2006) Salvage ra-

dioimmunotherapy with murine iodine-131-labeled antitenascin

monoclonal antibody 81C6 for patients with recurrent primary

and metastatic malignant brain tumors: phase II study results.

J Clin Oncol 24:115–122

274 J Neurooncol (2014) 116:267–274

123


Recommended