Antioxidative Enzymes, Carbonic Anhydrases and Claudins in Pediatric Brain Tumors

KRISTIINA NORDFORS Antioxidative Enzymes, Carbonic Anhydrases and Claudins in Pediatric Brain Tumors Prognostic and predictive value ACADEMIC DISSER...
Author: Lauren Haynes
3 downloads 2 Views 3MB Size
KRISTIINA NORDFORS

Antioxidative Enzymes, Carbonic Anhydrases and Claudins in Pediatric Brain Tumors Prognostic and predictive value

ACADEMIC DISSERTATION To be presented, with the permission of the board of the School of Medicine of the University of Tampere, for public discussion in the Jarmo Visakorpi Auditorium, of the Arvo Building, Lääkärinkatu 1, Tampere, on October 8th, 2011, at 12 o’clock.

UNIVERSITY OF TAMPERE

ACADEMIC DISSERTATION University of Tampere, School of Medicine Pirkanmaa Hospital District, Laboratory Centre Finland

Supervised by Docent Hannu Haapasalo University of Tampere Finland Professor Ylermi Soini University of Eastern Finland Finland

Reviewed by Professor Riitta Herva University of Oulu Finland Docent Olli Lohi University of Tampere Finland

Distribution Bookshop TAJU P.O. Box 617 33014 University of Tampere Finland

Tel. +358 40 190 9800 Fax +358 3 3551 7685 [email protected] www.uta.fi/taju http://granum.uta.fi

Cover design by Mikko Reinikka

Acta Universitatis Tamperensis 1648 ISBN 978-951-44-8546-6 (print) ISSN-L 1455-1616 ISSN 1455-1616

Tampereen Yliopistopaino Oy – Juvenes Print Tampere 2011

Acta Electronica Universitatis Tamperensis 1109 ISBN 978-951-44-8547-3 (pdf ) ISSN 1456-954X http://acta.uta.fi

Kristiina, Arvo, and Lea at Luosto 1986

To my inspirators and all the children with brain tumor 3

CONTENTS LIST OF ORIGINAL PUBLICATIONS

7

ABBREVIATIONS

8

ABSTRACT

10

TIIVISTELMÄ

12

1

INTRODUCTION

14

2

REVIEW OF THE LITERATURE

16

2.1. Pediatric brain tumors

16

2.1.1. Pilocytic astrocytoma

17

2.1.2. Ependymoma

18

2.1.3. Medulloblastoma

20

2.1.4. Primitive neuroectodermal tumor

22

2.2. Antioxidant enzymes

24

2.2.1. Manganese superoxide dismutase

26

2.2.2. Glutamate cysteine ligase, catalytic and regulatory subunit

27

2.2.3. Thioredoxin and thioredoxin reductase

28

2.2.4. Peroxiredoxins

30

2.2.4.1. Peroxiredoxin I

31

2.2.4.2. Peroxiredoxin II

32

2.2.4.3. Peroxiredoxin III

32

2.2.4.4. Peroxiredoxin IV

33

2.2.4.5. Peroxiredoxin V

33

2.2.4.6. Peroxiredoxin VI

34

2.3. Carbonic anhydrases

34

2.3.1. Carbonic anhydrase II

35

4

2.3.2. Carbonic anhydrase IX

36

2.3.3. Carbonic anhydrase XII

37

2.4. Claudins

39

2.4.1. Tight junctions

39

2.4.2. The blood-brain barrier

39

2.4.3. Claudin expression in normal tissue

40

2.4.4. Claudin expression in neoplasms

41

2.4.5. Epithelial-mesenchymal transition

44

3

AIMS OF THE STUDY

45

4

MATERIALS AND METHODS

46

4.1. Patients

46

5

4.1.1. Pilocytic astrocytomas

46

4.1.2. Ependymomas

46

4.1.3. Medulloblastomas and PNETs

47

4.2. Tumor tissue samples

47

4.3. Immunohistochemistry

48

4.3.1. Antioxidative enzymes and peroxiredoxins

48

4.3.2. Carbonic anhydrases

49

4.3.3. Claudins

49

4.3.4. Other immunohistochemistry and TUNEL

50

4.4. Histopathological features

51

4.5. Statistical methods

51

4.6. Ethics

51

RESULTS

52

5.1. Immunohistochemical expression

52

5

5.1.1. Antioxidative enzymes

52

5.1.2. Peroxiredoxins I-VI

52

5.1.3. Carbonic anhydrases II, IX, and XII

53

5.1.4. Claudins 2-5, 7, and 10

53

5.2. Clinicopathological features

55

5.2.1. AOEs and Prxs in pilocytic astrocytomas

55

5.2.2. AOEs in ependymomas

56

5.2.3. CAs in medulloblastomas and PNETs

56

5.2.4. CLDNs in ependymomas

57

5.3. Prognosis

6

57

5.3.1. Patients with pilocytic astrocytoma

57

5.3.2. Patients with ependymoma

58

5.3.3. Patients with MB or PNET

58

DISCUSSION

60

6.1. Current state of pediatric brain tumors

60

6.2. AOEs in pediatric brain tumors

60

6.3. Carbonic anhydrases in pediatric brain tumors

62

6.4. Claudins in pediatric brain tumors

63

6.5. The relationship between claudins and carbonic anhydrases

64

6.6. Limitations of the study

65

6.7. Future prospects

65

7

SUMMARY AND CONCLUSIONS

68

8

ACKNOWLEDGEMENTS

70

9

REFERENCES

72

10

ORIGINAL COMMUNICATIONS I-IV

110

6

LIST OF ORIGINAL PUBLICATIONS

This thesis is based on the following publications referred to in the text by their roman numerals.

I

Nordfors K, Haapasalo J, Helén P, Paetau A, Paljärvi L, Kalimo H, Kinnula VL, Soini Y and Haapasalo H (2007): Peroxiredoxins and antioxidant enzymes in pilocytic astrocytomas. Clin Neuropathol 26:210-218.

II Järvelä S*, Nordfors K*, Jansson M, Haapasalo J, Helén P, Paljärvi L, Kalimo H, Kinnula V, Soini Y and Haapasalo H (2008): Decreased expression of antioxidant enzymes is associated with aggressive features in ependymomas. J Neurooncol 90:283-291. (*shared first authorship).

III Nordfors K, Haapasalo J, Korja M, Niemelä A, Laine J, Parkkila AK, Pastorekova S, Pastorek J, Waheed A, Sly WS, Parkkila S and Haapasalo H (2010): The tumour-associated carbonic anhydrases CA II, CA IX and CA XII in a group of medulloblastomas and supratentorial primitive neuroectodermal tumours: an association of CA IX with poor prognosis. BMC Cancer 10:148.

IV Nordfors K, Haapasalo J, Sallinen P, Haapasalo H and Soini Y: Expression of claudins relates to tumour aggressivity, location and recurrence in ependymomas. (submitted)

Permission for the reproducing the original articles of this thesis has been given by the copywright owners.

7

ABBREVIATIONS AEC AIDS AML AOE AOP2 APC AXIN1 AXIN2 BBB Bcl-2 BRAF CA CAM CARPs CCTNNB1 cDNA CGNP CLDN C-MYC CNS CSF CT Cu/Zn-SOD CYS DNA D/N MB EC-SOD EMT -GC GEM GLCL GLCL-C GLCL-R GPX GSH GSSG GTR HER2 HIF HP iPNET LC/A MB MB MBEN MIB-1 MMP MN MnSOD

aminoethyl carbazole Aquired Immune Deficiency Syndrome acute myeloid leukemia antioxidative enzyme antioxidative protein 2 adenomatous polyposis coli axis inhibitor 1 axis inhibitor 2 blood-brain barrier B-cell lymphoma 2 v-raf murine sarcoma viral oncogene homolog B1 carbonic anhydrase cell adhesion molecule carbonic anhydrase-related protein cadherin- associated protein beta 1 complementary DNA cerebellar granule cell precursor claudin v-myc myelocytomatosis viral oncogene homolog central nervous system cerebro-spinal fluid computer tomography copper-zinc superoxide dismutase cysteine deoxyribonucleic acid desmoplasmic/nodular medulloblastoma extracellular superoxide dismutase epithelial-mesenchymal transition –glutamylcysteine genetically engineered mouse glutamate cysteine ligase gamma glutamyl cysteinyl synthetase catalytic subunit gamma glutamyl cysteinyl synthetase regulatory subunit glutatahione peroxidase glutathione oxidized glutathione gross-total resection Human Epidermal growth factor Receptor 2 hypoxia-inducible (transcription) factor Helicobacter pylori infratentorial primitive neuroectodermal tumor large cell/anaplastic medulloblastoma medulloblastoma medulloblastoma with extensive nodularity an antibody against Ki-67 matrix metalloproteinase carbonic anhydrase IX manganese superoxide dismutase

8

MRI mRNA NADPH NF1 NF-B NKEF-A NKEF-B N-MYC Nrf2 ORF6 PA PBS PG PKC PLA2 PNET PRC Prx PTCH1 PTEN RAS RCC RG ROS Shh SLUG SMOH SNAIL SOD SUFU sPNET TGR Tj TMA TMD TNF- TRAIL Trx TrxR TUNEL VEGF VEGFR vHL WHO ZEB1

magnetic resonance imaging messenger-ribonucleic acid nicotinamide adenine dinucleotide phosphate neurofibromatosis 1 nuclear factor kappa-light-chain-enhancer of activated B cells natural killer enhancing factor A natural killer enhancing factor B v-myc myelocytomatosis viral related oncogene, neuroblastoma derived nuclear factor 2 open reading frame 6 pilocytic astrocytoma phosphate-buffered saline proteoglycan protein kinase C phospholipase A2 primitive neuroectodermal tumor protein kinase C peroxiredoxin patched homolog 1 phosphatase and tensin homolog RAt Sarcoma renal cell carcinoma radial glia reactive oxygen species sonic hedgehog neural crest transcription factor, member of snail family smoothened homolog zinc finger transcription factor superoxide dismutase suppressor of fused homolog supratentorial primitive neuroectodermal tumor thioredoxin glutathione reductase tight junction tumor multitissue array transmembrane proteins with domain tumor necoris factor-alpha TNF-Related Apoptosis-Inducing Ligand thioredoxin thioredoxin reductase terminal deoxynucleotidyl transferase dUTP nick end labeling vascular endothelial growth factor VEGF receptor von Hippel-Lindau World Health Organization zinc finger E-box binding homeobox 1

9

ABSTRACT

Brain tumors are the second most common tumor type in children after leukemia. The outcome of the patients has become more favorable over the past few decades due to improved treatment modalities. Nowadays, the 5-year prognosis is from less than 60% to 90% depending on the tumor type. Nevertheless, the tumor itself and its treatment reduce quality of life, increase the risk of being handicapped, and raise expenses. Novel treatment modalities are under intense investigation, although a major breakthrough has yet to be discovered.

The aim of this thesis was to find new molecules to be used in the process of diagnosing and evaluating the predictivity, prognosis and follow-up of children with the most common pediatric brain tumors, including pilocytic astrocytomas, ependymomas, medulloblastomas (MBS), and primitive neuroectodermal tumors (PNETs). In this thesis, antioxidative enzymes were investigated in pilocytic astrocytomas and ependymomas, as well as peroxiredoxins in pilocytic astrocytomas. In addition, the role of carbonic anhydrases was studied in medulloblastomas and PNETs. Finally, claudins were analysed in ependymomas.

The first study analysed the antioxidative enzymes (AOEs), including manganese superoxide dismutase (MnSOD), gamma glutamyl cysteinyl synthetase catalytic and regulatory subunits (GLCL-C, GLCL-R), thioredoxin (Trx), thioredoxin reductase (TrxR) and peroxiredoxins (Prx) IVI in a series of 105 pilocytic astrocytomas. All of them were expressed in pilocytic astrocytomas, suggesting that oxidative damage and consequent defence take place during the progression of the tumors. AOEs correlated with the degenerative features and angiogenesis, possibly associating with reactive oxygen species-derived cellular damage. Moreover, the expression of the AOEs was associated with each other in terms of concurrent activation of the enzymes. With the exception of MnSOD, a strong expression of AOEs was generally associated with higher cell proliferation. Prx VI seemed to have a positive association with a longer recurrence-free interval.

The second study investigated the relationship between AOE (MnSOD, GLCL-C, GLCL-R, Trx, TrxR) expression and clinicopathological features in 67 ependymal tumors. Most of the tumors expressed AOEs. Lower GLCL-C and GLCL-R expression was associated with higher tumor grade. MnSOD, GLCL-C and TrxR expressions were significantly higher in tumors located in the spinal cord compared with those in the brain. Interestingly, decreased expression of Trx predicted worse

10

outcome for the patients. This finding may have clinical relevance when planning treatment modalities and follow-up for patients.

The aim of the third study was to analyse the expression of carbonic anhydrases (CAs) II, IX, and XII in a set of 39 medulloblastomas and PNETs. Interestingly, CA II was expressed in both the neovessel endothelium and tumour cell cytoplasm. CA IX was mainly expressed in the tumor cells located in perinecrotic areas. CA XII showed the most homogenous distribution within the tumors. Importantly, CA IX expression predicted poor prognosis in both univariate and multivariate analyses. CA IX has been previously found to be a promising target molecule for anticancer treatment in other tumors. The results suggest that this could also be the case for medulloblastomas and PNETs.

In the fourth study, expression of claudins (CLDNs) 2-5, 7, and 10 was investigated in a set of 61 ependymomas. According to the results, all CLDNs except for CLDN4 were expressed in these tumors. CLDN5 was related to more aggressive tumors compared with CLDN2 and 10. Tumors expressing these two claudins displayed a better degree of differentiation and a better prognosis. There were also differences in the expression of claudins associated with location of the tumor and between primary and recurrent tumors, CLDNs 3 and 5 were more often found in the cerebrum than in other sites and CLDN7 in primary tumors compared with recurrent ones. Evidently claudins influence the growth and differentiation in ependymomas.

In summary, the studied antioxidative enzymes, peroxiredoxins, carbonic anhydrases, and claudins were expressed in the most common pediatric brain tumors, including pilocytic astrocytomas, ependymomas, medulloblastomas, and PNETs. Prx VI was associated with longer recurrence-free interval in patients with pilocytic astrocytoma, whereas decreased Trx expression predicted worse prognosis of patients with ependymoma. CA IX correlated with worse outcome in patients with medulloblastoma or PNET. Claudins had no significant association with prognosis, nevertheless CLDN5 was related to more aggressive ependymomas.

11

TIIVISTELMÄ

Aivokasvaimet ovat lasten toiseksi yleisin kasvaintyyppi leukemian jälkeen. Erityisesti hoitojen kehityttyä lasten ennuste on parantunut selvästi viime vuosikymmenten aikana. Nykypäivänä 5vuotisennuste vaihtelee kasvaimesta riippuen alle 60%:sta 90%:iin. Tauti ja hoidot aiheuttavat kuitenkin elämänlaadun heikkenemistä, vammautumisia, sekä myös kustannuksia. Uusia hoitokeinoja tutkitaan jatkuvasti, mutta selviä läpimurtoja ei ole toistaiseksi saavutettu. Väitöskirjani tavoitteena oli etsiä uusia molekyylejä, joita voitaisiin hyödyntää lasten yleisimpien aivokasvainten diagnostiikassa, ennusteen arvioimisessa sekä seurannassa. Kirjassa läpikäydään antioksidatiivisten entsyymien esiintymistä pilosyyttisissä astrosytoomissa sekä ependymoomissa. Lisäksi

väitöskirjassani

käsitellään

peroksiredoksiineja

pilosyyttisissä

astrosytoomissa,

hiilihappoanhydraaseja medulloblastoomissa ja primitiivisissä neuroektodermaalisissa tuumoreissa (PNET:ssa), sekä klaudiineja ependymoomissa.

Ensimmäisessä osajulkaisussa tutkittiin antioksidatiivisten entsyymien (AOE; MnSOD, GLCL-C, GLCL-R, Trx, TrxR), sekä peroxiredoksiinien (Prx I-VI) esiintymistä 105 pilosyyttisessä astrosytoomassa. Kaikki ilmentyivät laajasti kyseisissä kasvaimissa. Tämä viittaa oksidatiivisen vaurion ja sen korjausmekanismien osallisuuteen pilosyyttisen astrosytooman kehittymisessä. Lisäksi AOE:t ilmentyivät kasvaimissa samanaikaisesti viitaten mahdolliseen yhteiseen aktivoitumiseen. Kaikilla AOE:lla oli yhteys korkeaan proliferaatioasteeseen, lukuun ottamatta MnSOD:ia. Prx VI oli tilastollisesti merkittävä tekijä arvioitaessa kasvaimen uusiutumista.

Toisessa osajulkaisussa tutkittiin AOE:n (MnSOD, GLCL-C, GLCL-R, Trx, TrxR) yhteyttä 67 ependymooman kliinispatologisiin muuttujiin. Useimmat kasvaimet ilmensivät entsyymejä. GLCLC:n ja GLCL-R:n vähäisyys oli yhteydessä korkeampaan gradukseen. MnSOD, GLCL-C ja TrxR värjäytyminen oli voimakkaampaa selkäytimen kuin aivojen ependymoomissa. Kiinnostava löytö oli Trx:n vähäisyyden yhteys huonoon ennusteeseen. Tällä havainnolla voi olla kliinistä merkitystä potilaiden hoidon ja seurannan suunnittelussa.

Kolmas osatyö käsitteli hiilihappoanhydraasien (CA) medulloblastoomassa

ja

PNET:ssa.

CA

II

esiintyi

II,

IX ja XII esiintymistä 39

uudisverisuonten

endoteelissä

sekä

sytoplasmassa, kun taas CA IX värjäytyi voimakkaimmin nekroottisten alueiden reunoilla. CA XII esiintyvyys oli tasaisempaa. CA IX oli huonon ennusteen merkki sekä yksimuuttuja, -että monimuuttujamalleissa. CA IX on osoittautunut lupaavaksi syövänhoidon kohdemolekyyliksi 12

muissa kasvaimissa ja osatyön perusteella näin voisi olla myös medulloblastoomien ja PNET:n kohdalla.

Neljännessä osajulkaisussa tutkittiin klaudiinien (CLDN 2-5,7 ja 10) esiintymistä 61 ependymoomassa. Lukuunottamatta CLDN4, kaikki klaudiinit esiintyivät ependymoomissa. CLDN5:llä oli yhteys kasvaimen aggressiivisuuteen, kun taas CLDN2:llä ja 10:llä oli trendi parempaan erilaistumiseen ja ennusteeseen. Klaudiinit ilmentyivät erilailla kasvaimen sijainnista riippuen; CLDN3 ja 5 värjäytyivät voimakkaammin isojen aivojen ependymoomissa. CLDN7 värjäytyvyys oli voimakkaampaa primaarikasvaimissa. Tuloksista voi päätellä, että klaudiinit vaikuttavat ependymoomien kasvuun ja erilaistumiseen.

Tutkitut antioksidatiiviset entsyymit, mukaanluettuna peroxiredoksiinit, sekä hiilihappoanhydraasit ja klaudiinit esiintyivät lasten yleisimmissä aivokasvaimissa (pilosyyttisissä astrosytoomissa, ependymoomissa, medulloblastoomissa sekä PNET:ssa). Prx VI oli yhteys pilosyyttisen astrosytooman uusiutumiseen, kun taas Trx:n väheneminen ennusti ependymooma-potilaiden huonompaa ennustetta. CA IX sen sijaan oli huonon ennusteen merkki medulloblastooma ja PNET –potilailla. Klaudiineilla ei ollut yhteyttä potilaiden ennusteeseen, mutta CLDN5 assosioitui ependymooman aggressiivisuuteen.

13

1

INTRODUCTION

Pediatric brain tumors are the most common solid tumors and the second most common tumors after leukemia in children. Although the prognosis has improved over the past few decades, children still have severe side effects from the treatment; they may be handicapped, and 5-year survival varies from less than 60% to 90% (Louis et al. 2007).

There are several types of pediatric brain tumor. Pilocytic astrocytoma (PA), is a grade I glial tumor arising mostly in the cerebellum of children (Ohgaki and Kleihues 2005). It is histologically benign but may present a clinical challenge to neurosurgeons due to its location. Nevertheless, the prognosis remains good and the 5-year survival rate is 80-90% (Watson et al. 2001, Mueller and Chang 2009).

Another tumor entity is grade I-III ependymoma, which originates from ependymal cells or their stem cells. The typical location is the 4th ventricle, though ependymomas may occur at any site along the ventricular system and in the spinal cord (Schiffer et al. 1991, Prayson 1999). The outcome of the patients depends mainly on age, tumor location, extent of resection, and histopathological grading (Jayawickreme et al. 1995, Ernestus et al. 1996, Horn et al. 1999, Louis et al. 2007). Children with intracranial ependymoma have a 50% 5-year progression-free survival rate (Robertson et al. 1998, Venkatramani et al. 2011).

The most common malignant brain tumor in children is grade IV medulloblastoma (MB). Embryonal MB locates in the vermis and 4th ventricle, and it may metastasize via cerebro-spinal fluid (Louis et al. 2007). The 5-year survival rate for patients with MB is 60-70%. The improvement compared to previous survival rate is mainly due to better treatment modalities (Ellison et al. 2003). Central nervous system (CNS) primitive neuroectodermal tumors (PNETs) are a group of tumors recently reclassified (Louis et al. 2007). PNET is a grade IV embryonal tumor with early onset and aggressive behavior. The prognosis is worse than with medulloblastoma (Geyer et al. 2005) and younger children have the worst outcome (Geyer et al. 1994).

The standard of care for brain tumors is neurosurgical gross total resection (GTR) and it is applied where possible. Adjuvant therapy, including chemo- and radiotherapy, is being used in selected cases (Mueller and Chang 2009). In addition, novel treatment modalities and prognosticators have been under intense investigation. Nevertheless, comprehensive and clinically valid tools have yet to 14

be developed. The blood-brain barrier (BBB) is a challenge for anticancer treatment.

Most

molecules are not capable of penetrating the BBB. In order to penetrate, the molecule should be electrically neutral, lipid-soluble, and small. The neural tissue is very sensitive to damage and this is another challenge for anticancer treatment.

Accurate knowledge of tumor biology is the basis for studying and diagnosing brain tumors. Genetic analyses give novel information and may lead to more individual treatments. This is unfortunately still not cost-effective for clinical use. The present study was conducted to establish new tools for clinical screening, diagnosis, as well as prognostic markers and methods for monitoring and predicting the outcome for patients with pediatric brain tumors. The study results may facilitate future research strategies for the field of brain tumors in children.

15

2

REVIEW OF THE LITERATURE

2.1. PEDIATRIC BRAIN TUMORS

Brain tumors are the second most common tumor type in children after leukemia. Typical pediatric type brain tumors are pilocytic astrocytomas, ependymomas, medulloblastomas and PNETs (Louis et al. 2007). Although most of the tumors can occur in any age group, the aforementioned tumors are typically seen in children or young adults (Figure 1) (Kieran et al. 2010). The biology of the tumors differs to some extent between age groups and thus, the following text concentrates on pediatric brain tumors.

Figure 1. Incidence of pilocytic astrocytoma, ependymoma, medulloblastoma, and PNET in different age groups. Modified from Kieran et al. (2010). 10 9

Incidence per 1 million

8 7 6 5

Pilocytic astrocytoma

4

Ependymoma

3

Medulloblastoma/PNET

2 1 0 0-14

15-19

20-34

Age (years)

16

35-44

2.1.1. Pilocytic astrocytoma Pilocytic astrocytoma (PA) is the most common astrocytoma of children, comprising 21% and 16% of all CNS tumors for age groups 0-14 years and 15-19 years, respectively (Central Brain Tumor Registry 2006). In children, the tumor arises mostly in the cerebellum (Ohgaki and Kleihues 2005), though other sites include optic chiasm, hypothalamus, thalamus, basal ganglia, cerebral hemispheres, brain steam and spinal cord (Louis et al. 2007). Although the tumor corresponds to World Health Organization (WHO) grade I and the 5-year overall survival is good, 80-90%, patients with pilocytic astrocytoma may have poor outcome owing to localisation (Watson et al. 2001, Mueller and Chang 2009) (Table 1). Patients can suffer from headache, visual loss, hydrocephalia, hemiparesis or may even die of the disease. Most frequently, pilocytic astrocytoma of patients less than 20 years old presents as clumsiness, worsening headache, nausea and vomiting.

The radiological diagnosis of pilocytic astrocytomas is made by computer tomography (CT) or magnetic resonance imging (MRI), in which it is present as a circumscribed and contrast enhancing tumor (Lee et al. 1989, Fulham et al. 1993). The tumor has typical cysts, which are an important detail when evaluating the tumor grade (Palma et al. 1983).

The histolopathology of pilocytic astrocytoma is heterogenous. The cells are bipolar piloid with Rosenthal fibers and multipolar with microcysts and granular bodies. Pilocytic astrocytomas have a low mitotic index, nuclei are hyperchromatic and pleomorphic. Although pilocytic astrocytomas often have vascular proliferation, the tumor is still of a benign type. Endothelial proliferation and necrosis, typically seen in high-grade astrocytomas, are rarely seen in PAs (Louis et al. 2007). In the literature, there are few examples of pilocytic astrocytoma undergoing malignant transformation, but the tumor should not be confused with glioblastoma because it is then an anaplastic pilocytic astrocytoma (Dirks et al. 1994, Tomlinson et al. 1994).

Pilocytic astrocytoma is associated with neurofibromatosis 1(NF1). Patients with NF1 usually have PA in the optic nerve and thus, about 30% of patients with optic nerve PAs have NF1. NF1associated tumors grow slowly or remain stable (Hoyt and Baghdassarian 1969). Although the cause of PA remains uncertain, people with NF1, Li-Fraumeni syndrome and prior radiation to the brain carry a higher risk of PA development (Mueller and Chang 2009).

Patients with PA undergo radical resection where possible. The extent of resection is strongly associated with survival (Laws et al. 1984). New operation techniques have enabled the most 17

eloquent brain areas accessible. Recurrences do occur and are often a reflection of cyst reformation. Nevertheless, recurrence-free survival is greater than 95% at 10 and 20 years in patients with radical resection (Watson et al. 2001).

If complete resection is possible, it is rare for patients to undergo additional adjuvant therapy, unless there is evidence of recurrence or progression from MRI (Table 1). Typical genetic alterations seen in diffusively infiltrating astrocytomas are rarely seen in PAs (Cheng et al. 2000) and thus, similar future treatment modalities can not be used. Previous studies show that there is trisomy of chromosome 5 and 7 or gain of 1q (Jones et al. 2006). In addition, several studies have shown a mitogen-activated protein kinase pathway activation in PAs and duplication of the v-raf murine sarcoma viral oncogene homolog B1 (BRAF) gene locus (Pfister et al. 2008).

2.1.2. Ependymoma Ependymoma is mainly a tumor of children and young adults, though it may occur in all age groups (Central Brain Tumor Registry 2006). The age distribution depends on the histological type and location, younger patients having mostly an infratentorial tumour and 30-40 year olds mostly a spinal tumor (Waldron and Tihan 2003). Otherwise, the tumor may occur at any site in the CNS, but mostly in the fourth ventricle, in the spinal cord and the lateral and third ventricles (Schiffer et al. 1991, Prayson 1999). One third of ependymomas are localised supratentorially and two thirds infratentorially (Zacharoulis and Moreno 2009) (Table 1).

Most ependymomas are considered to be WHO histological grade II. Other ependymomas include anaplastic ependymoma (grade III) and rare entities: grade I myxopapillary ependymoma and subependymoma, grade I. Grade II ependymoma consists of four variants, such as cellular ependymoma (the most common), papillary, clear cell, and tanycytic ependymoma. The tumor cells are usually round or oval and may contain eosinophilic cytoplasmic granules and dense chromatin material. Grade III ependymoma has a higher proliferation and mitotic index, though distinction of grade II from grade III can be rather difficult (Louis et al. 2007) (Table 1).

Symptoms and signs are dependent on location. Typically the symptoms are caused by mechanical compression of the cerebral fluid circulation leading to hydrocephalus, headache, nausea, vomiting, dizziness and, with very young children, head enlargement (Duncan and Hoffman 1995).

18

The tumor is initially diagnosed with gadolinium-enhanced MRI, in which ependymoma is well circumscribed and shows varying degrees of contrast enhancement (Furie and Provenzale 1995). About half of the patients have multifocal calcification, and some of these tumors are hemorrhagic (Zacharoulis and Moreno 2009).

The molecular basis of ependymoma remains uncertain. Chromosome 22 mutations are the most frequent finding, and about 30% of cases have monosomy or translocation in this chromosome (Reni et al. 2007). Additionally, anaplastic ependymoma has an association with gain in chromosome 1q and this chromosomal change is also correlated to a worse outcome (Rand et al. 2008). Comparative genomic hybridisation has revealed a variable number of genetic imbalances. Added to this, abnormal Notch and Sonic Hedgehog (Shh) signaling have been reported in ependymomas (Modena et al. 2006). Similarly to other brain tumors, the pathogenesis of ependymomas may include neural cancer stem cells (Taylor et al. 2005).

Surgical resection is the standard of care for patients with ependymoma. Gross total resection (GTR) can be applied in about 40-60% of cases, and is obviously more common with a supratentorial location (Zacharoulis and Moreno 2009). Even children with a metastatic tumor benefit from GTR (Zacharoulis et al. 2008). Chemotherapy has only a minor role in the treatment of ependymomas. Evans et al. (1996) have shown in a randomised study that adding vincristine and lomustine after radiotherapy does not improve survival. Infants may be the only patient group who may benefit from chemotherapy. In contrast, focal radiotherapy is an important treatment modality for children with ependymoma. Approximately half of the patients will experience relapse, typically locally in the first two years, though late recurrences are also seen (Zacharoulis and Moreno 2009). The most common treatment for patients with a relapse is re-operation and this increases progression-free survival (Vinchon et al. 2005). Adjuvant chemotherapy has a modest effect on survival (Zacharoulis and Moreno 2009) whereas radiotherapy is beneficial for patients with relapsed ependymoma (Combs et al. 2006) (Table 1).

The prognosis of the patients relies mainly on four different clinical features. First, tumor location is the main feature determing patient survival. Posterior fossa tumors are usually more aggressive than supratentorial tumors. Ependymoma in the spinal cord tend to have late recurrences, but still have a better prognosis compared to cerebral neoplasms (Ernestus et al. 1996). Second, the extent of the resection is an independent prognosticator as well (Jayawickreme et al. 1995). Third, there are many histopathological features indicating patient outcome. Patients have a worse prognosis 19

when ependymoma shows anaplasia, e.g. tumor cells are less differentiated and show a high mitotic index and enhanced proliferation (Schiffer and Giordana 1998, Korshunov et al. 2004, Kurt et al. 2006). Fourth, children under three years of age seem to have a worse outcome compared to the elderly (Horn et al. 1999).

2.1.3. Medulloblastoma Medulloblastoma (MB) is a malignant, WHO grade IV brain tumor occuring mainly in children (Arseni and Ciurea 1981). The peak age is seven years, and 70% of the patients are less than 16 years of age (Roberts et al. 1991). There is a male predominance (65%). Medulloblastomas are usually located in the cerebellar vermis and the fourth ventricle. Older patients usually have desmoplastic/nodular subtypes, which locate mainly in the cerebellar hemipheres (Louis et al. 2007) (Table 1). In addition, desmoplastic MB occurs at a high frequency among infants (Ellison 2010).

According to the WHO classification MB is separated into classic tumor and four variants: desmoplasmic/nodular (D/N), MB with extensive nodularity (MBEN), anaplastic and large cell MB. Classic MB is the most common medulloblastoma. Large cell and anaplastic MB are aggressive tumors, whereas MBENs and D/N MBs in infants have better outcome than classic MB. (Louis et al. 2007) D/N and MBEN typically have nodules of differentiated neurocytic cells and internodular desmoplasia (Giangaspero et al. 1999). Large cell MB contains groups of uniform large cells with round nuclei and a single nucleus. Both large cell MB and anaplastic MB show a high mitotic activity and apoptosis. Anaplastic MB is dominated by nuclear pleomorphism (Eberhart et al. 2002). Many studies combine D/N MB and MBEN as desmoplastic tumors, and large cell and anaplastic MB as large cell/anaplastic (LC/A) tumors (Table 1). Classic medulloblastoma has monotous small cells and the nuclei may be either round or oval. Some tumors show rosettes and palisades (Ellison 2010).

The molecular pathology of MBs is under extensive research. There is evidence of abnormalities in Shh pathway through mutations in patched homolog 1 (PTCH1), smoothened homolog (SMOH) and suppressor of fused homolog (SUFU) genes in 25% of medulloblastomas. This works as a stimulator to cerebellar granule cell precursors (CGNPs) and Purkinje cells to release Shh during CNS development, a phenomenon which has been shown in several genetically engineered mouse (GEM) models. Another pathway is the Wnt pathway comprising 15% of MBs. In this pathway, there are mutations in cadherin- associated protein), beta 1 (CTNNB1), adenomatous polyposis coli. (APC), and axis inhibitor (AXIN)1/2. The pathway produces nuclear accumulation of -catenin, 20

which acts as a transcriptional activator (Ellison 2010). In addition to the two signaling pathways, there are two non-Shh/Wnt subgroups, which are associated with up-regulation of specific gene classes, but not with aberrant activation of signaling pathways (Northcott et al. 2011). The nonShh/Wnt tumors account for 60% of medulloblastomas (Ellison 2010). Isochromosome 17q (i17q) is the most frequent structural aberration in medulloblastomas, found in 30–40% of cases (Northcott et al. 2009). V-myc myelocytomatosis viral related oncogene, neuroblastoma derived (N-MYC) gene amplification has also been identified in up to 10% of medulloblastoma specimens and, like vmyc myelocytomatosis viral oncogene homolog (C-MYC), is often found in tumors with large cell/anaplastic features (Aldosari et al. 2002).

Patients with medulloblastoma often have mechanical obstruction of cerebro-spinal fluid (CFS-) flow, and this is usually the reason for typical symptoms. These include increased intracranial pressure, headache, vomiting and nausea. Patients may also have excessive lethargy and ataxia. CTscans or MRI reveal a solid, intensely contrast-enhancing mass (Louis et al. 2007).

The prognosis of young patients has increased over the past few decades, and nowadays the 5-year survival rate is 60-70%. This improvement is mainly due to better treatment modalities. The most important treatment is surgical resection, but perioperative chemo- and radiation therapy are also applied (Ellison et al. 2003). MBs are very radiosensitive tumors, so children over three years of age have radiation therapy. Side effects of the treatment have led to a reduction in the amount of radiation (Deutsch et al. 1996). The problem is that lowering the radiation dose without adding chemotherapy leads to a worse outcome and thus, chemotherapy is a standard choice for children with MB (Mueller and Chang 2009) (Table 1). The patients are divided into low and high risk (age 75% of tumor cells showing positivity. In addition to the researcher, two experienced pathologists analyzed the staining results, and a consensus meeting was held in the case of a disagreement.

4.3.2. Carbonic anhydrases Parkkila et al. (1993) have produced and characterised rabbit antiserum against human CA II. The previously described monoclonal antibody M75, recognising the N-terminal domain of human CA IX, was used in the CA IX immunostaining procedure (Pastorekova et al. 1992, Chrastina et al. 2003). Rabbit anti-human CA XII antiserum against the secretory form of CA XII has been previously characterised (Karhumaa et al. 2000) and was used in this study. The detailed staining protocol is described in study III.

The staining reactivities for CA II, CA IX and CA XII were scored from multitissue- blocks on a scale from 0 to 3 as follows: 0 = no reaction, 1 = weak reaction (< 10% positive cells), 2 = moderate reaction (10-30% positive cells), 3 = strong reaction (>30% positive cells). Due to the sample size, staining results were categorised into two groups: negative staining was considered as CA-negative and weak, moderate and strong staining were considered as CA-positive.

4.3.3. Claudins The primary antibodies for the detection of claudins 2-5, 7, and 10 in formalin-fixed paraffinembedded tissues were purchased from Zymed Laboratories Inc (South San Francisco, CA). The antibodies included polyclonal rabbit anti-claudin 2 (clone JAY.8), polyclonal rabbit anti-claudin 3 (clone Z23.JM), monoclonal mouse anti-claudin 4 (clone 3E2C1), monoclonal mouse anti-claudin 5 (clone 4C3C2), polyclonal rabbit anti-claudin 7 (clone ZMD.241) and polyclonal anti-rabbit claudin 10 (Ca N:o 38-8400).

Immunostaining of claudins was carried out as follows. The microarray sections were first heated in a microwave oven in 10 mM citrate buffer (pH 6.0) for 10 minutes. After a 60-minute incubation with the primary antibody (dilution 1:50 for anti-claudin 1, 3, 4, 5 and 7, 1:100 for claudin 10), a biotinylated secondary anti-rabbit or anti-mouse antibody and Histostain-SP kit (Zymed Laboratoris Inc) were used. The bound antibodies were demonstrated with diaminobenzidine as a chromogen. 49

The sections were then lightly counterstained with haematoxylin and mounted with Eukitt (Kindler, Freiburg, Germany). Positive controls included non-neoplastic kidney, breast, skin and liver samples. Non-immune rabbit or mouse serum were used as substitutes for the primary antibodies to act as the negative control.

Two neuropathologists analyzed the staining results, and a consensus meeting was held in the case of a disagreement. Membrane bound immunoreactivity was considered significant, although cytoplasmic expression was also detected to be present. The immunoexpression of claudins was assessed as follows; negative = < 5% of cells positive, weak = 5-25 % of cells positive, moderate = 25-75 % of cells positive, strong = 75-100 % of cells positive. Due to the sample size, the tumors were recorded as CLDN-negative or CLDN-positive (from weak to strong staining) for statistical analyses.

4.3.4. Other immunohistochemistry and TUNEL Analysis of cell proliferation was carried out using a mouse monoclonal antibody MIB-1 (Ki-67 antigen, dilution 1:40, Immunotech, S.A. Marseille, France). The tissue sections were counterstained with methyl green. The proliferative activity was reported as a percentage of nuclei with positive immunoreaction. The analysis of Ki-67/MIB-1 positive nuclei in tissues was evaluated quantitatively using a computer-assisted image analysis system (CAS-200 TM Software). Immunohistochemical analysis for p53 status was performed as described earlier (Haapasalo et al. 2003). Apoptosis was detected using ApopTagTM In Situ Apoptosis Detection Kits (Oncor Inc., Gaithersburg, MD) as described previously (Haapasalo et al. 1999).

The mouse monoclonal TWIST (ab50887, Abcam, Cambridge, UK) and ZEB1 (clone 416A7H10, GenWay, San Diego, CA, USA) antibodies at a 1:500 dilution with the microarray sections were incubated overnight at 4°C. The slides were then stained using a standard avidin-biotin-enhanced immunoperoxidase technique (ABC Vectastain Elite Kit, Vector Laboratories, Burlingame, CA, USA). Diaminobenzidine tetrahydrocloride (DAP, in phosphate-buffered saline) (Sigma, St. Louis, MO, USA) was used as chromogen. The sections were counterstained with Mayer's haematoxylin, washed, dehydrated, cleared and mounted with Depex (BDH, Poole, UK). Ovarian tumor tissue, known to be positive for TWIST and ZEB1 expression, was used as a positive control. Nuclear immunoreactivity for TWIST and ZEB1 was considered significant. Tumors with widespread staining (over 5% positivity) were recorded as TWIST/ZEB1-positive.

50

4.4. Histopathological features The presence of the following histological features was recorded for further statistical analyses in case of ependymomas: atypia, endothelial proliferation, necrosis, mitosis, hypercellularity, and pseudorosettes, true rosettes, and papillarity. In pilocytic astrocytomas and ependymomas, calcification and myxoid change were recorded.

4.5. Statistical methods All statistical analyses were performed using SPSS for Windows 11.0 (Chicago, IL). The significance of the associations was defined using the chi-square test, the Mann-Whitney test and the Kruskal-Wallis test. A log-rank test, Kaplan-Meier curves and Cox multivariate regression analysis were used in the survival analysis.

4.6. Ethics The study designs for all the original publications were approved by the Ethics committee of Tampere University Hospital and the National Authority for Medicolegal Affairs.

51

5

RESULTS

5.1. Immunohistochemical expression

5.1.1. Antioxidative enzymes Antioxidative enzymes, including MnSOD, Trx, TrxR, GLCL-C, and GLCL-R, were studied in 105 pilocytic astrocytomas (96 primary and 9 recurrent tumors) and in 67 ependymomas (46 primary and 21 recurrent tumors). Pilocytic astrocytomas showed mainly cytoplasmic AOE immunostaining pattern.

In primary ependymomas, AOE immunoreactivity was as follows. 87% showed immunoreactivity for MnSOD. Positive immunoreactivity for GLCL-C was found in 74% of the cases and 89% for GLCL-R. Trx immunoreactivity was seen in 72%. 54% of primary tumors were TrxR immunoreactive (Table 6).

The correlations between AOEs in ependymomas were as follows (unpublished data): negative MnSOD expression was associated with TrxR negativity, (p = 0.017, chi-square test). Similarly, GLCL-C negativity correlated with negative expressions of GLCL-R, Trx, and TrxR (p = 0.002, p = 0.023, p < 0.001, chi-square test, respectively). GLCL-R was more often negative when there was negative TrxR expression (p = 0.005, chi-square test). As expected, Trx and TrxR were expressed simultaneously (p = 0.003, chi-square test).

5.1.2. Peroxiredoxins I-VI Peroxiredoxins (Prxs) were analysed in 105 pilocytic astrocytomas. In the tumor cells the immunostaining was cytoplasmic, but Prxs I, II, IV, V and VI also showed variable nuclear positivity. For Prx I, 98 % of cases were positive). All tumors were Prx II positive and also Prx III. Most (92%) of all tumors expressed positivity for Prx IV, (13 % strongly, 49 % moderately, 30 % weakly) and 87% were positive for Prx V. Prx VI expression was found in 97 %, of all tumors (Table 6).

In the further statistical analysis, the tumors were recorded Prx-negative (no and weak immunostaining) or Prx-positive (moderate and strong immunopositivity). Co-expression was found between different Prxs as follows. Prx I was associated with Prx II, Prx IV, and Prx VI (p < 0.001, p = 0.010, p < 0.001, chi-square test, respectively). Prx II was associated with Prx IV and Prx 52

VI (p = 0.002, p < 0.001, chi-square test, respectively). Similarly, Prx III was associated with Prx IV (p = 0.004, chi-square test). Prx IV was associated with all other Prxs (Prx V, p = 0.007; Prx VI, p = 0.010, chi-square test, respectively).

5.1.3. Carbonic anhydrases II, IX, and XII Carbonic anhydrases were investigated in 39 medulloblastomas and PNETs. CA II was found in the endothelium of neovessels and the cytoplasm of MB/PNET cells. Endothelial CA II expression was detected in 49% of all tumors, whereas, cytoplasmic CA II was found in 73% of the cases. CA IX and CA XII were less frequently expressed than CA II in the tumor samples: CA IX positivity was found in 23% of tumors, and 11% of the tumors were positive for CA XII (Table 6). Similar to previous studies, CA IX expression was linked to necrosis, while CA XII was more homogenously stained. In further analysis, the tumors were recoded as CA-negative and CA-positive (weak, moderate and strong staining combined). The co-expression of CA II, IX and XII in the subgroups of MBs and PNETs or in the total tumor material did not reach statistical significance (chi-square test).

5.1.4. Claudins 2-5, 7, and 10 Claudins were analysed in a group of 61 ependymoma samples. CLDNs were rather widely expressed, except for CLDN4 which was not expressed in our tumor material. CLDN2 -positivity was found in forty-two percent of all), and 23% of the tumors were positive for CLDN3. For CLDN5, 26% of ependymomas showed positive immunostaining). Most (81%) of the tumors expressed CLDN7, and 11% of cases were positive for CLDN10 (Table 6).

There was an association between negative CLDN3 and negative CLDN5 (p = 0.010, chi-square test). None of the other claudins had correlations with each other.

53

Table 6. Expression of peroxiredoxins, antioxidative enzymes, claudins, and carbonic anhydrases in pediatric brain tumors.

Intensity of expression Pilocytic astrocytomas Prx I Prx II Prx III Prx IV Prx V Prx VI Ependymomas MnSOD GLCL-C GLCL-R Trx TrxR CLDN2 CLDN3 CLDN5 CLDN7 CLDN10 Medulloblastomas/PNETs CA II endothelial CA II cytoplasmic CA IX CA XII

negative

weak

moderate

strong

2% 0% 0% 82% 13% 3%

3% 76% 27% 30% 35% 15%

11% 20% 47% 49% 23% 37%

84% 4% 26% 13% 29% 45%

13% 26% 11% 28% 46% 58% 77% 74% 19% 89%

46% 30.5% 41% 35% 17% 14% 9% 5% 45% 7%

30% 17.5% 33% 33% 26% 16% 7% 5% 31% 4%

11% 26% 15% 4% 11% 12% 7% 16% 5% 0%

51% 27% 77% 89%

6% 24% 7% 5%

11% 38% 13% 3%

32% 11% 3% 3%

54

5.2. Clinicopathological features Various clinicopathological features were tested in statistical analysis. The features were divided as patient dependent, including patient age and sex, or tumor dependent (location, and molecular pathological features).

5.2.1. AOEs and Prxs in pilocytic astrocytomas The pilocytic astrocytomas of the older patients had more intense staining for MnSOD and GLCLC (p = 0.022 and p = 0.010, respectively, Mann-Whitney test). In addition, recurrent tumors had a significantly lower MnSOD staining pattern (p = 0.019, Mann-Whitney test). No such differences were seen as regards Prxs. Additionally, there was no association between location of tumors and enzyme expression (chi-square test).

Expression of Prx II, Prx III, Prx IV, GLCL-C and TrxR were associated with vascular pathology in pilocytic astrocytomas. Positive Prx II and Prx III expressions had an association with higher endothelial proliferation (p = 0.046, p = 0.023, chi-square test, respectively). In addition, cells with less immunopositivity for GLCL-C also had less vascular hyalinisation (p = 0.013, chi-square test), and similarly lower levels of TrxR had less perivascular lymphocytes (p = 0.006, chi-square test). We analysed AOEs’ relationship with degenerative features and found that Prx I, Prx II and TrxR had significant correlations: Prx I, Prx II, and TrxR were more positive in tumors with cystic pattern (p < 0.001, p = 0.002, p = 0.027, chi-square test, respectively). Expression of GLCL-R and TrxR correlated significantly with an aggressive tumor growth pattern. Positivity for TrxR was associated with a higher atypia rate (p = 0.004, chi-square test), whereas tumors with negative staining pattern for GLCL-R were less necrotic (p = 0.008, chi-square test).

Proliferation index (Ki-67/MIB-1) was also studied in pilocytic astrocytomas. We found that Prx VI and TrxR positive tumors had a higher proliferation rate (Prx VI: negative tumors, 1.8 ± 3.1 (mean±sd), positive tumors 3.0 ± 3.1, p = 0.037; TrxR: negative tumors, 2.2 ± 3.1, positive tumors, 3.5 ± 3.0, p = 0.028, Mann-Whitney test, respectively). However, MnSOD -positivity was associated with lower proliferative activity (negative tumors, 3.3 ± 3.2, positive tumous 2.2 ± 2.9, p = 0.039, Mann-Whitney test). AOE -status and p53 immunopositivity or terminal deoxynucleotidyl transferase dUTP nick end labeling (terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), apoptosis) positivity (Mann-Whitney test) were not associated with AOEs.

55

5.2.2. AOEs in ependymomas In ependymomas, GLCL-C and GLCL-R expression was linked to tumor aggressivity in such a way that the expression decreased significantly as the grade increased (p = 0.047 and 0.049, chi-square test). When the recurrent tumors were included in the analysis, GLCL-C expression was even more significantly different (p = 0.026, chi-square test).

When tumor site (spinal vs. brain) and AOE expression were compared, tumors were divided into AOE-negative and AOE-positive groups; negative and faint staining was considered as a negative group and moderate and strong staining as a positive group. In the total tumor material, all AOEs were even more intensely stained in spinal tumors than in the intracranial tumors. This difference was statistically significant for MnSOD, GLCL-C and TrxR expression (p = 0.044, 0.046 and 0.004, respectively, chi-square test).

We did not find associations between AOEs and tumor proliferation rate (Ki-67/MIB-1), nor with p53. In contrast, B-cell lymphoma 2 (Bcl-2) –positivity correlated significantly with MnSOD, GLCL-C, Trx and TrxR expression (p = 0.049, p = 0.037, p = 0.024 and p = 0.022, respectively, chi-square test). Though p53 had no significant association to other clinicopathological features, higher proliferation correlated to higher tumor grade (p = 0.033, Kruskal-Wallis test). In addition, lower grade tumors were linked to Bcl-2 positivity (p < 0.001, chi-square test), higher patient age (p = 0.029, Kruskal-Wallis test) and spinal tumor location (p < 0.001, chi-square test).

5.2.3. CAs in medulloblastomas and PNETs CA II, IX and XII expression were also analysed in corcondace with various clinical features and molecular markers. Proliferation (Ki-67/MIB-1), apoptosis (chi-square and Mann-Whitney test) or expression of Bcl-2, p53 or c-erbB-2 were not associated with CAs in any of the groups except for the correlation between positive c-erbB-2 and positive CA IX expression in PNETs (p = 0.047, chisquare test). Interestingly, the tumors of young patients had more CA XII-positivity (total material p < 0.001, MBs p < 0.001, chi-square test). CA IX was also positivitely associated with female gender (total material p = 0.048, MBs p = 0.023, chi-square test). There was no significant difference in the expression of CAs between primary and recurrent tumors in any of the groups (chi-square test). Moreover, there were no correlation between tumor type (MBs/PNETs) and CA intensity (chi-square test).

56

5.2.4. CLDNs in ependymomas The association between claudins and several clinicopathological features were analysed in ependymomas. We found that different claudins had particular effects on tumor behavior. When tumor grade and claudins were compared, we found that tumors with high grade were often CLDN5 immunopositive (p = 0.049, chi-square test), whereas CLDN10 positive ependymomas were more often low grade tumors (p = 0.039, chi-square test). Ependymomas with different locations also had different CLDN expression. CLDN5 and CLDN3 were more often found in the cerebrum than in the cerebellum or the spinal cord (p = 0.036, p = 0.007, chi-square test, respectively). When primary and recurrent tumors were compared with their claudin expression, only CLDN7 had significant association, as CLDN7 immunoreactivity was more often found in primary than in recurred ependymomas (p = 0.041, chi-square test).

Typical histopathological features of ependymomas were also linked to claudin expression. This was the case especially with CLDN5, which was more often positively expressed in tumors with increased nuclear atypia, endothelial proliferation, mitotic rate and hypercellularity (p = 0.007, p = 0.018, p = 0.041, p = 0.010, respectively, chi-square test). CLDN5 positive tumors also showed higher Ki-67/MIB-1 cell proliferation rate than CLDN5 negative tumors (p = 0.015, Mann-Whitney test). The immunoexpression of the other claudins was not associated with histological features or cell proliferation index of ependymomas.

When EMT associated transcription factors ZEB1 and TWIST were analysed in ependymomas we found that tumors positive for ZEB1 were often negative for CLDN2 (p = 0.031, chi-square test). Negative expression of TWIST was also associated with negative expression of CLDN5 and CLDN10 (p = 0.013, p = 0.017, chi-square test, respectively). Neither ZEB1 nor TWIST was associated with tumor grade, cell proliferation rate, or histological features.

5.3. Prognosis

5.3.1. Patients with pilocytic astrocytoma All 96 patients with primary pilocytic astrocytomas were included in the survival analysis. None of the studied AOEs or Prxs had an association with overall survival, whereas tumors immunopositive for Prx VI seemed to be associated with significantly better recurrence-free survival when compared with immunonegative tumors (p = 0.032, log-rank test) (Table 7). However, when patient age, Ki-67/MIB-1, location (cerebellum versus other location) and Prx VI –immunopositivity were 57

included in multivariate analysis, none of these factors independently predicted recurrence-free survival (Cox multivariate analysis).

5.3.2. Patients with ependymoma Patients with primary ependymoma (N = 46 in study II, and N = 44 in study IV) were included in the survival analysis. Among the studied AOEs, only Trx had statistical significance for patient survival; tumors that did not express Trx had worse survival in follow-up than patients whose tumors were Trx-positive (p = 0.045, log-rank test). Curiously, we found that with adults (age >19 years), Trx acted as a prognosticator of better outcome and all whose tumors expressed Trx were alive at the end of the follow-up period (p = 0.011, log-rank test). Younger patients did not have a similar effect. In Cox multivariate analysis, Trx was the only independent prognosticator (Odds ratio 7.30, 95% confidence interval 0.93-55.56, p = 0.059), whereas WHO grade, location (spinal vs. brain) and patient age (child vs. adult) were not included in the Cox model (Table 7).

When prognosis of the patients with primary ependymoma (N = 44) was analysed with a log rank test, we found that CLDNs were not associated with patient prognosis. Nevertheless, it seems that patients whose tumors express CLDN2, 5, or 10 may have better clinical outcome than those with CLDN3 or 7 positivity. Interestingly, all the patients with CLDN10 positive tumors or CLDN7 negative tumors were alive at the end of follow-up. TWIST and ZEB1 had no effect on patient outcome.

5.3.3. Patients with MB or PNET All 35 patients with primary MB/PNET were included in the survival analysis. CA IX-positivity was a marker of worse outcome in patients with MB/PNET (all tumors p = 0.041, MBs p = 0.030, PNETs p = n.s.; log-rank test). In addition, patients with medulloblastoma had worse prognosis when their tumors showed CA XII positivity (p = 0.010, log-rank test). When the prognostic value of CAs was tested in the multivariate analysis, we included the following prognostic indicators: patient age, Ki-67/MIB-1 proliferation index, apoptosis index and expression of p53, c-erbB-2 and Bcl-2. In addition, the histopathological type (MB vs. supratentorial PNET), CA II, CA IX and CA XII were used in the analysis. Interestingly in our material, only expression of CA IX (odds ratio 4.31; 95% confidence interval (CI) 1.31 - 14.11; p = 0.016) and the apoptosis index (odds ratio 3.29; 95% CI 1.05 - 10.31, p = 0.041) were independent prognostic factors (Table 7). The expression of neither CA II nor CA XII showed a significant association with survival.

58

Table 7. Association between the studied enzymes and the prognosis of the patients with pediatric brain tumors. Pilocytic astrocytoma Prx VI+  recurrences  Ependymoma Trx-  prognosis  Medulloblastoma/PNET CA IX+  prognosis  CA XII+  prognosis  (medulloblastoma) Apoptosis+  prognosis  +/- = positive/negative immunostainig  = better prognosis  = less recurrences/worse prognosis

59

6

DISCUSSION

6.1. Current state of pediatric brain tumors Pediatric cancers are still the main cause of death in children aged 1-14 years in the UK and Finland (Gatta et al. 2005, Statistics Finland 2003-2009). Brain tumors are the second largest tumor group in children after leukemia and the current 5-year survival rate for patients with brain tumor is approximately 60-70% in Nordic countries (Gatta et al. 2005), including Finland (Pokhrel and Hakulinen 2009).

The evolving brain is vulnerable. Even though patients survive, the brain tumor itself and its treatments can cause side effects. The majority of children with a brain tumor live an active life with only minor disabilities. Unfortunately, about a half of the patients do have neurological motor disabilities, and even more patients have an abnormal neurological status. Supratentorial tumors, tumor reoperations, shunt revisions and chemotherapy increase the risk of these problems (Lannering et al. 1990, Ilveskoski et al. 1996, Reimers et al. 2003). There is only a limited scope for improvement with conventional chemotherapy and thus, there is an urgent need for novel, therapeutic agents for these patients.

6.2. AOEs in pediatric brain tumors Our aim was to study the expression of different AOEs in pilocytic astrocytomas and ependymomas. In addition, we analysed the expression of related Prxs in pilocytic astrocytomas.

AOEs and Prxs were widely expressed in pilocytic astrocytomas. According to our study, Prxs and other AOEs were commonly expressed simultaneously. This suggests that the neoplastic tissue suffers from a constant oxidative load, leading to simultaneous up-regulation of these enzymes. Coexpression and a relationship between Prxs have also been observed in previous studies of other neoplasms (Kinnula et al. 2002, Karihtala et al. 2003). An increased oxidative load may lead to degenerative features and cellular destruction in tumor tissue. Prx enzymes and other antioxidant enzymes were usually associated with an elevated proliferation rate, but MnSOD seemed to depress proliferative activity. This is in line with its putative tumor suppressor function, which is further supported by the fact that in the recurrences the expression of MnSOD was significantly decreased. Isolated over-expression of MnSOD decreases the growth, proliferation and invasion of cultured malignant cells (Bravard et al. 1992, St Clair et al. 1992, Yan et al. 1996, Zhong et al. 1997, reviewed by Kinnula and Crapo 2004). Some AOEs have a role as tumor suppressors, and thus, we 60

included proliferation index (Ki-67/MIB-1), the apoptosis regulating Bcl-2 and p53 in the studied variables.

Although there was no association between apoptosis rate and AOEs, histopathologically degenerative and regressive features were often associated with Prx expression. Cystic degeneration with Rosenthal fibres (often seen in pilocytic astrocytomas) and calcification implies cell death, which may be a sign of previous hypoxic periods. Elevated levels of AOEs in pilocytic astrocytomas with such degenerative features indicate the tendency of these tumors to suffer from oxidative stress and increased defence against ROS- mediated tissue damage. Prx II, Prx III, Prx IV, GLCL-c and TrxR were associated with vascular pathology, e.g. endothelial proliferation as a sign of angiogenesis. Hypoxia and ROS induce HIF-alpha, which stimulates angiogenic factors such as VEGF (Kinnula and Crapo 2004). In diffuse astrocytomas it is most probably hypoxia that induces the expression of VEGF in tumor cells. This binds to VEGF receptors (VEGFRs) in endothelial cells, leading to endothelial vascular proliferation. In pilocytic astrocytomas endothelial vascular proliferation is also a common feature and could be a consequence of ROS and AOE induction, but without such a sinister implication as in diffuse astrocytomas.

Furthermore, many AOEs, such as MnSOD and Prxs, increase chemo- and radio-resistance of tumor cells. Contrary to diffuse astrocytomas, pilocytic astrocytomas are treated by surgery alone. This might partly explain the differences in AOEs/Prxs and association with tumor behavior. Even though Prx VI was associated with higher cell proliferation, it seemed to be associated with significantly better recurrence-free survival. This might be due to an association between Prx VI expression and young patient age.

The majority of 67 ependymomas expressed MnSOD, GLCL-C, GLCL-R, Trx and TrxR. However, the intensity of immunoreactivity had a decreasing trend as the tumor grade increased. This is the opposite phenomenon to diffusively infiltrating astrocytomas or oligodendrogliomas (Haapasalo et al. 2003, Järvelä et al. 2006). Lack of antioxidant protection would render tumor cells more vulnerable to DNA damage, which during the development and growth of higher-grade ependymomas might be expressed as higher aneuploidy and higher malignancy.

Furthermore, AOE expression (for MnSOD, GLCL-C and TrxR) depended on the location of the tumor, as spinal tumors showed more intense staining patterns than tumors of the brain. The reason for this remains obscure, but may indicate either biologic differences between the tumors of these 61

two sites or alternatively, there may be some fundamental differences in the oxidative milieu between different compartments of the central nervous system. Furthermore, grade I myxopapillary ependymomas differ from grade II and III tumors in their typical conus- cauda filum terminale location which makes it possible to resect the tumor radically.

Positive AOE (except for GLCL-R) expression had correlation with a well-known inhibitor of apoptosis, Bcl-2. ROS induce apoptosis and AOEs counteract apoptosis and cell damage by neutralizing ROS. It seems interesting that those AOEs that affect apoptotic activity have upstream regulatory steps to that of Bcl-2. One theoretical explanation would be that when oxidative stress is below a critical threshold, AOEs are expressed to protect the cell from irreversible damage and immediate apoptosis is not appropriate. Finally, we discovered that there is an association between higher Trx expression and longer survival, as discussed later.

6.3. Carbonic anhydrases in pediatric brain tumors We found that several MBs and supratentorial PNETs in a group of 39 tumor samples express the CA isozymes CA II, CA IX and CA XII. The expression of CA II, CA IX and CA XII in the normal nervous system has been investigated in several previous studies. The localization of CA II is well documented in the normal human oligodendrocytes (Kumpulainen et al. 1983). Based on our previous studies, CA IX is not present in the normal human brain, except for choroids plexus (Haapasalo et al. 2006). CA XII mRNA expression has been shown to be very weak in the human brain by RT-PCR analysis (Haapasalo et al. 2008). In the mouse, immunohistochemistry has shown that CA XII is in the choroid plexus (Kallio et al. 2006). A recent study by Chiche et al. (2009) provided clear evidence that both CA IX and CA XII are functionally involved in tumor growth, including in renal cell cancer. In vivo experiments showed that CA9 gene silencing alone led to a 40% reduction in xenograft tumor volume, and the silencing of both CA9 and CA12 resulted in an 85% reduction in tumor volume. In medulloblastomas and PNETs we found that CA IX was found in the perinecrotic areas of the tumors whenever necrosis was present. Similar findings have been reported in astrocytomas (Haapasalo et al. 2006). Because necrosis is an uncommon feature and is not considered to be a significant prognostic factor in MBs, the induction of CA IX in MBs/PNETs may also involve hypoxia-independent mechanisms. Interestingly, CA II was, once again, found in the endothelium of neovessels. Thus, CA II may play an important functional role in tumor metabolism. The expression of CA XII in the tumor cells was associated with patient age, as previously reported for the expression of CA XII in patients with diffuse astrocytomas (Haapasalo et al. 2008). These findings reflect the fact that patient age is a significant factor that contributes to 62

carcinogenesis by several mechanisms, including CA expression, and that tumor phenotypes are different depending on the age of the patient. The expression of CA IX and apoptotic activity were associated with a poor prognosis, as discussed later. Although in the total tumor material CA II and CA XII did not reach statistical significance for use as prognostic indicators, CA II had a similar trend to that of CA IX. Furthermore, CA XII showed a significant correlation with survival in MBs.

6.4. Claudins in pediatric brain tumors In our material, claudins 2,3,5,7, and 10 were widely expressed in 61 ependymomas. CLDN4 was not expressed in our material. CLDNs seemed to be even more highly expressed than in the previous study (Hewitt et al. 2006). We found differences associated with location of the tumor and between primary and recurrent tumors in the expression of claudins. CLDNs 3 and 5 were more often found in tumors of the cerebrum than in the cerebellum or the spinal cord. Such regional variation in their expression may originate from the expression pattern of parental ependymal cells in the liquor space which may vary as has been shown in other tissues (Rahner et al. 2001, KiuchiSaishin et al. 2002). In addition, primary tumors showed more intense CLDN7 staining patterns compared with recurrent ones. Our results showed claudin 7 downregulation in the progression of ependymoma, but curiously, all patients with CLDN7 negative tumors survived compared to the positive group. CLDN 7 is a member of the family which is abundantly present in epithelial tumors (Soini 2005). However, depending on tumor type, claudin 7 expression may be over- or underexpressed. Downregulation in malignant melanoma, head and neck cancer and in breast cancer is reasonable considering the disruption of tight junctions leading to loss of cohesion and invasiveness (Krämer et al. 2000, Kominsky et al. 2003). Interestingly, CLDN7 is upregulated in hepatocellular carcinoma, ovarian epithelial cell carcinoma, prostate carcinoma or renal chromophobe carcinoma (Singh et al. 2010).

CLDN5 immunoreactivity in ependymomas associated with increased nuclear atypia, endothelial proliferation, mitotic rate and hypercellularity, higher proliferation, and tumor grade. Our study indicates that CLDN5 tends to be related to more aggressive tumors. In contrast, CLDN2 and 10 tend to display a better degree of differentiation and a better prognosis. Claudin 2 tended to be expressed in ependymomas with a better prognosis, even though no significant association was found. Claudin 2 is commonly present in epithelial tumors and, as with claudins 7 and 10, its presence has been associated with leakiness of the tight junctions (Soini 2005, Krause et al. 2008). Our study showed claudin 10 to be expressed in 11 % of cases, and expression tended to be located

63

in low grade tumors with a better prognosis. Evidently claudins influence the growth of and differentiation in ependymomas.

The expression of claudins is partly regulated by epithelial-mesenchymal transition (EMT) associated transcription factors, such as snail1, slug, ZEB1 or TWIST (Martinez-Estrada et al. 2006). An inverse association between ZEB1 and claudins 1 and 2, and TWIST and claudin 5 has been shown in lung carcinoma (Merikallio et al. 2011). ZEB1 was inversely associated with claudin 2 in ependymoma, whereas claudins 5 and 10 showed a positive correlation with TWIST. Indeed, ependymomas contained a high frequency of ZEB1 expression compared to lung carcinomas, for instance (Merikallio et al. 2011). This may be due to the importance of ZEB1 in neural development. In mice, activation of ZEB1 mRNA expression is found in cells of the periventricular area (Yen et al. 2001). Thus, ZEB1 might serve other functions in ependymoma, not necessarily related to invasion type features like EMT which is actually a phenomenon related to epithelial tumors. TWIST expression was found in ependymomas with a lesser frequency. In glioblastoma cells, TWIST expression has been found to be related to a more aggressive behaviour and mesenchymal change (Mikheeva et al. 2010). Such a discrepancy may be due to the special nature of the ependymal cells and neoplasias which do not straightforwardly correspond to epithelial tumors.

6.5. The relationship between claudins and carbonic anhydrases Previous studies show that both claudins and carbonic anhydrases participate in the formation of tight junctions and the BBB. CA II is necessary for human vasculogenesis and formation of the BBB (Kida et al. 2006). Like claudins, CA IX is a cell adhesion molecule (CAM) (Závada et al. 2000) and seems to play a role in intercellular adhesion with its proteoglycan-like region (Svastová et al. 2003). CA IX can perturb E-cadherin-mediated cell-cell adhesion via interaction with β– catenin and potentially contribute to tumor invasion (Svastová et al. 2003).

Furthermore, CA IX is known to be hypoxia-inducible (Wykoff et al. 2000). Hypoxia has also been shown to regulate the barrier function of neural blood vessels by reducing the expression of claudin 5 in endothelial cells (Koto et al. 2007). This could indigate that claudins and carbonic anhydrases may be connected in multiple ways. AOEs and related Prxs are induced in hyperoxic conditions and protect cells from ROS. (Halliwell 1991, Rabilloud et al. 2002). Hypoxia is known to be one of the causes of treatment failure and poor outcome in a variety of adult malignancies by increasing resistance to radiotherapy and to cytotoxic agents (Teicher 1994, Brown et al. 2006). In addition, 64

hypoxia increases invasion, angiogenesis and metastasis (Tatum et al. 2006). Resistance to druginduced apoptosis in hypoxia has been found in pediatric tumors, e.g. neuroblastoma (Das et al. 2005, Hussein et al. 2006). Tyrosine kinase inhibitor, AZD 2171, has been studied initially in children with tumors including medulloblastoma, and one third of the patients showed growth delay of the tumor (Maris et al. 2008). Interestingly, CA IX inhibitors have been under intense investigation and promising results have been published in tumors like breast cancer (Robertson et al. 2004). In children, possible side-effects may be more severe and the exact biology of hypoxia and its clinical relevance in childhood tumors is still unclear. Thus, further studies will be needed before novel agents concerning hypoxia can be introduced into pediatric oncology.

6.6. Limitations of the study Small sample size is probably the major limitation of the study. Fortunately, children with brain tumors are not very common in Finland. This has necessitated certain procedures in our study, including recording different diagnostic and prognostic markers in statistical analyses. Additionally, there were disparate treatment modalities in the different hospitals, heterogeneous age variation and a rather long time interval between the first and last patients brain tumor operations.

Technical limitations included complex staining methods with numerous laboratory steps. It would be of interest to stain the tumors with different antibodies and verify the staining results of the thesis. Although staining protocols and TMA system have been widely and repeatedly used, one could utilize different laboratory steps and thus study the repeatability.

Despite small sample size, the thesis reached many statistically significant results concerning essential features of brain tumors, such as tumor behavior, diagnostics, and prognosis. In order to assess the clinical value of our findings, larger clinical trials with patients receiving standardised brain tumor treatments are required. This may, however, be a challenge since treatments are expected to develop when new data based on genetic profiling of individual patients is available.

6.7. Future prospects It is clear that brain tumors of children are rare, and thus collaboration is needed between reseachers. Others have found interesting and novel possible diagnostic and prognostic tools for patients with pilocytic astrocytoma, ependymoma, medulloblastoma, or PNET. The main question is which tool is clinically valid and cost-effective for clinical use.

65

According to our study, Prx VI is a marker of recurrence-free survival in pilocytic astrocytomas. This is probably a result of the fact that Prx VI over-expression in pilocytic astrocytomas without the bias produced by radio- or chemotherapy decreases ROS activity and in this way might abrogate genomic instability and tumor progression. The tumor is histologically benign but the clinical outcome may sometimes be poor and thus, Prx VI negativity could be used to define patients with higher risk and could indicate that these patients should have a closer follow-up. There are only a few prognosticators found for patients with pilocytic astrocytoma (Haapasalo et al. 1999). In earlier studies, univariate statistical analyses found that partial resection, older patient age, and histology (Haapasalo et al. 1999), especially pilomyxoid tumor variant, (Fernandez et al. 2003) were associated with a worse prognosis.

According to our study, decreased Trx-expression in ependymomas is associated with worse outcome. Interestingly, a similar association between high AOE expression and better prognosis has been shown in breast cancer where peroxiredoxins III and IV expression was associated with better survival (Karihtala et al. 2003). Furthermore, in oligodendroglial or diffuse astrocytic brain tumors, high Trx was associated with a worse prognosis of the patients (Haapasalo et al. 2003, Järvelä et al. 2006), whereas in ependymomas it was associated with an improved prognosis. Such a phenomenon could also be ascribed to the protective function of Trx and other AOEs against oxidant damage which, in the case of AOE expressing ependymomas, would mean a higher genetic stability and less aggressive behavior. In addition, there are differences between the treatment of different tumor types and this might be one reason for the opposite relationship between AOEs and tumor behavior. Accordingly, AOEs have been shown to have an effect on anti-cancer drug resistance (Tew 1994, Kinnula and Crapo 2004). The stem cell/progenitor cell origin of ependymal cells is also different from that of astrocytes and oligodendrocytes (Linskey and Gilbert 1995), which might be one reason for differences, as well. Embryonic radial glia (RG) are neural progenitor cells that are likely to be the source of ependymomas independent of patient age. Astroglial cells with functional and molecular characteristics of RG persist in the supraventricular zone of the lateral ventricles and possibly the spinal cord, suggesting that some RG give rise to adult neural stem cells (Merkle et al. 2004, Barry and McDermott 2005). RG-derived stem cells may be the cells of origin of adult ependymomas.

In our study on medulloblastomas and PNETs, we found that CA II was, once again, found in the endothelium of neovessels. Thus, CA II may play an important functional role in tumor metabolism. CA II has been found to be a target molecule for dendritic cell therapy in melanoma patients 66

(Yoshiura et al. 2005). Further studies are, therefore, clearly warranted to evaluate the role of CA II as a possible therapeutic target not only in melanoma but also in other forms of cancer, including MBs/PNETs.

CA IX was found to be of prognostic value in medulloblastomas in our material. Similar findings have been reported previously in other tumors as well. The clear division of CA IX expression in normal and neoplastic cells makes CA IX a promising diagnostic and prognostic tool in various tumors. CA IX is associated with higher grade, necrosis, and worse outcome in breast tumors (Chia et al. 2001, Wykoff et al. 2001). A similar effect is seen in head and neck cancer (Beasley et al. 2001, Koukourakis et al. 2001), in bladder cancer (Hoskin et al. 2003), in cervix carcinoma (Loncaster et al. 2001), and in in RCC (Bui et al. 2003 and 2004). CA IX seems to have several inductors. It has also been shown that higher CA IX expression is associated with a more favourable overall survival in some tumors, such as RCC and in acute myeloid leukemia (AML). In RCC, the CA IX induction is associated with VHL-mutation and not with hypoxia as in most brain tumors (Patard et al. 2008). In AML the association is thought to be involved with immune system and Tcell response (Greiner et al. 2006).

Claudins did not reach statistically significant correlations with patient prognosis, but there were interesting trends between CLDNs and outcome of patients with ependymoma. Previous studies have shown that CLDNs have potential as prognosticators as well. Due to high specificity of CLDN expression in cancer, it has been suggested that claudins may represent useful molecular markers for many different cancers, such as CLDN3 in ovarian cancer (Lu et al. 2004) and CLDN10 as an independant prognosticator for recurrence of hepatocellular carcinoma (Cheung et al. 2005). CLDN5 expression is high in brain cancer in vascular endothelial cells, and thus may represent a target for antiangiogenic therapy (Hewitt et al. 2006). In addition, Nitta et al. (2003) have shown that knockout of CLDN5 results in a selective increase in paracellular permeability of small molecules, and thus makes CLDN5 a possible target for the development of drugs for this purpose.

In the future, children diagnosed with brain tumor will be more accurately stratified based on a combination of clinical variables and molecular profiles. Improved risk stratification will enable individualised therapies, which could be a combination of conventional treatment modalities and novel, targeted therapeutic approaches. These changes will hopefully result in improved survival without detriment to the quality of life.

67

7

SUMMARY AND CONCLUSIONS

1. Altogether eleven different antioxidative enzymes were studied in pilocytic astrocytomas. All AOEs (MnSOD, GLCL-C, GLCL-R, Trx, TrxR, and Prx I-VI) were expressed in pilocytic astrocytomas suggesting that oxidative damage and consequent defence take place during the progression of the tumors. AOEs correlated with degenerative features and angiogenesis, possibly associating with reactive oxygen species derived cellular damage. Moreover, the expression of the AOEs was associated with each other in terms of concurrent activation of the enzymes. With the exception of manganese superoxide dismutase (MnSOD), a strong expression of AOEs was generally associated with higher cell proliferation. Prx VI seemed to have a positive association with a longer recurrence-free interval.

2. When five AOEs (MnSOD, GLCL-C, GLCL-R, Trx, TrxR) were studied in ependymomas, some AOEs seemed to be associated with tumor grade and location in ependymomas. Lower GLCL-C and GLCL-R expression was associated with higher tumor grade. MnSOD, GLCLC and TrxR expressions were significantly higher in tumors located in the spinal cord compared to those in the brain. Interestingly, decreased expression of Trx predicted worse outcome for the patients. This finding may have clinical relevance when planning the treatment modalities and follow-up for the patients.

3. Carbonic anhydrases II, IX, and XII were widely expressed in medulloblastomas and PNETs. CA XII was associated with young patient age. Interestingly, CA IX was found to be of prognostic importance in medulloblastomas and PNETs. Previous studies have shown that CA IX is an attractive target molecule for anticancer treatment. Additional studies will be needed to analyse whether CA IX could also be used in practice in the case of medulloblastomas and PNETs.

68

4. Claudins 2, 3, 5, 7, and 10 were widely expressed in ependymomas. CLDN5 tends to be related to more aggressive tumors compared with CLDN2 and 10, which tend to display a better degree of differentiation and a better prognosis. There were also differences associated location of the tumor and between primary and recurrent tumors in the expression of claudins; CLDNs 3 and 5 were more often found in the cerebrum than in other sites and CLDN7 in primary tumors compared with recurrent ones. Evidently claudins influence the growth of and differentiation in ependymomas.

69

8

ACKNOWLEDGEMENTS

The present study was carried out at the Department of Pathology, University Hospital of Tampere. My deep gratitude goes to the entire staff working there and the professor of the department, Timo Paavonen, M.D., Ph.D.

I am greatly indebt to my supervisors, Docent Hannu Haapasalo, M.D., Ph.D. for his consistent encouragement in the field of science, and Professor Ylermi Soini, M.D., Ph.D., the most efficient and reliable researcher. They both have introduced me to scientific thinking and working. Many thanks for your support and patience.

I thank Professor Seppo Parkkila, M.D., Ph.D. and Docent Anna-Kaisa Parkkila, M.D., Ph.D. for their work as the members of the supervisory committee and collaborators. They have both given me wise advice and new ideas.

I appreciate the time, constructive criticism and suggestions given by Professor Riitta Herva, M.D., Ph.D, and Docent Olli Lohi, M.D., Ph.D, the official referees of this manuscript.

I wish to thank my collaborators, Professors Hannu Kalimo, M.D., Ph.D, Vuokko Kinnula, M.D., Ph.D, Silvia Pastorekova, M.D., Ph.D, Jaromir Pastorek, M.D., Ph.D, Abdul Waheed, M.D., Ph.D, and William S. Sly, M.D., Ph.D. for their valuable co-work. I sincerely thank Docents Leo Paljärvi, M.D., Ph.D, Pauli Helén, M.D., Ph.D, Jukka Laine, M.D., Ph.D, and Anders Paetau, M.D., Ph.D for their expertise. I would like to thank also Sally Järvelä, M.D., Ph.D, Pauli Sallinen, M.D., Ph.D, Miia Jansson M.D., and Anssi Niemelä, M.D. Especially I acknowledge Miikka Korja, M.D., Ph.D, who has taught me that only the sky is the limit.

Special thanks to Mrs Reija Randen, Mrs Eila Pohjola, Ms Aija Parkkinen, Mrs Aulikki Lehmunen, Mrs Riitta Koivisto, Mr Jorma Lampinen, Mrs Päivi Koukkula and Mr Manu Tuovinen for skillful, technical assistance. I also thank Docent Mikko Arola, M.D., Ph.D., for his expertise on pediatric oncology in the third manuscript. I thank Nick Bolton, Ph.D. for revising the first study.Prakash Oommen, M.D. and Mike Nelson, Ph.D. are acknowledged for their help in revising the language of the second manuscript. The Proofreading247 Team is acknowledged for revising this thesis.

70

My parents Liisa and Heimo I would like to thank for their love. They have encouraged me to study medicine and kept my feet on the ground when needed. I am grateful to my sisters Hannele and Katri and their families for support, kindness and being interested in my work. I warmly thank all my friends for great moments and relaxing company.

This study was financially supported by the Nona and Kullervo Väre Foundation, the Finnish Medical Society, Competitive Research Funding of the Tampere Medical Research Fund of Tampere University Hospital (Grants 9L067, 9J013 and 9M010), by the Irja Karvonen Cancer Trust, by the Orion-Farmos Research Foundation, the EVO foundation of the Northern Savo district, the Finnish Cancer Society, the anti-tuberculous association of Finland, EU 6th Framework programme (DeZnIT), and the Kevo funding of Kuopio University Hospital. They are all acknowledged.

And the last and the dearest thanks go to my husband, Joonas. Never have I met such a person, who is so enthusiastic about research. Joonas has pushed me forward when needed and supported me every step of the way. There are no words to describe the gratitude. Thank you for your patience and love.

Tampere, August 2011

Kristiina Nordfors

71

9

REFERENCES

Abbott NJ, Ronnback L and Hansson E (2006): Astrocyte-endothelial interactions at the bloodbrain barrier. Nat Rev Neurosci 7:41–53.

Aldosari N, Bigner SH, Burger PC, Becker L, Kepner JL, Friedman HS and McLendon RE (2002): MYCC and MYCN oncogene amplification in medulloblastoma. A fluorescence in situ hybridization study on paraffin sections from the Children’s Oncology Group. Arch Pathol Lab Med 126:540–544.

Ando-Akatsuka Y, Saitou M, Hirase T, Kishi M, Sakakibara A, Itoh M, Yonemura S, Furuse M and Tsukita S (1996): Interspecies diversity of the occludin sequence: cDNA cloning of human, mouse, dog, and rat-kangaroo homologues. J Cell Biol 133:43-47.

Araki M, Nanri H, Ejima K, Murasato Y, Fujiwara T, Nakashima Y and Ikeda M (1999): Antioxidant function of the mitochondrial protein SP-22 in the cardiovascular system. J Biol Chem 274:2271-2278.

Arnér ES and Holmgren A (2000): Physiological functions of thioredoxin and thioredoxin reductase. Eur J Biochem 267:6102-6109.

Arnér ES and Holmgren A (2006): The thioredoxin system in cancer. Semin Cancer Biol 16:420426.

Arthur FE, Shivers RR and Bowman PD (1987): Astrocyte-mediated induction of tight junctions in brain capillary endothelium: an efficient in vitro model. Brain Res 433:155-159.

Arseni C and Ciurea AV (1981): Statistical survey of 276 cases of medulloblastoma (1935-1978). Acta Neurochir 57:159-162.

72

Bailey HH, Gipp JJ, Ripple M, Wilding G and Mulcahy RT (1992): Increase in gammaglutamylcysteine synthetase activity and steady-state messenger RNA levels in melphalan-resistant DU-145 human prostate carcinoma cells expressing elevated glutathione levels. Cancer Res 52:5115–5118.

Barry D and McDermott K (2005): Differentiation of radial glia from radial precursor cells and transformation into astrocytes in the developing rat spinal cord. Glia 50:187–197.

Bartosová M, Parkkila S, Pohlodek K, Karttunen TJ, Galbavý S, Mucha V, Harris AL, Pastorek J and Pastoreková S (2002): Expression of carbonic anhydrase IX in breast is associated with malignant tissues and is related to overexpression of c-erbB2. J Pathol 197:314-321.

Basu A, Banerjee H, Rojas H, Martinez SR, Roy S, Jia Z, Lilly MB, De León M and Casiano CA (2010): Differential expression of peroxiredoxins in prostate cancer: Consistent upregulation of PRDX3 and PRDX4. Prostate Oct 28. [Epub ahead of print].

Beasley NJ, Wykoff CC, Watson PH, Leek R, Turley H, Gatter K, Pastorek J, Cox GJ, Ratcliffe P and Harris AL (2001): Carbonic anhydrase IX, an endogenous hypoxia marker, expression in head and neck squamous cell carcinoma and its relationship to hypoxia, necrosis, and microvessel density. Cancer Res 61:5262-5267.

Bello IO, Vilen ST, Niinimaa A, Kantola S, Soini Y and Salo T (2008): Expression of claudins 1, 4, 5, and 7 and occludin, and relationship with prognosis in squamous cell carcinoma of the tongue. Hum Pathol 39:1212-1220.

Bertini R, Howard OM, Dong HF, Oppenheim JJ, Bizzarri C, Sergi R, Caselli G, Pagliei S, Romines B, Wilshire JA, Mengozzi M, Nakamura H, Yodoi J, Pekkari K, Gurunath R, Holmgren A, Herzenberg LA, Herzenberg LA and Ghezzi P (1999): Thioredoxin, a redox enzyme released in infection and inflammation, is a unique chemoattractant for neutrophils, monocytes, and T cells. J Exp Med 189:1783-1789.

Bravard A, Sabatier L, Hoffschir F, Ricoul M, Luccioni C and Dutrillaux B (1992): SOD2: a new type of tumor-suppressor gene? Int J Cancer 51:476-480.

73

Brown LM, Cowen RL, Debray C, Eustace A, Erler JT, Sheppard FC, Parker CA, Stratford IJ and Williams KJ (2006): Reversing hypoxic cell chemoresistance in vitro using genetic and small molecule approaches targeting hypoxia inducible factor-1. Mol Pharmacol 69:411-418.

Bui MH, Seligson D, Han KR, Pantuck AJ, Dorey FJ, Huang Y, Horvath S, Leibovich BC, Chopra S, Liao SY, Stanbridge E, Lerman MI, Palotie A, Figlin RA and Belldegrun AS (2003): Carbonic anhydrase IX is an independent predictor of survival in advanced renal clear cell carcinoma: implications for prognosis and therapy. Clin Cancer Res 9:802-811.

Bui MH, Visapaa H, Seligson D, Kim H, Han KR, Huang Y, Horvath S, Stanbridge EJ, Palotie A, Figlin RA and Belldegrun AS (2004): Prognostic value of carbonic anhydrase IX and KI67 as predictors of survival for renal clear cell carcinoma. J Urol 171:2461-2466.

Carmeliet P, Dor Y, Herbert JM, Fukumura D, Brusselmans K, Dewerchin M, Neeman M, Bono F, Abramovitch R, Maxwell P, Koch CJ, Ratcliffe P, Moons L, Jain RK, Collen D, Keshert E and Keshet E (1998): Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 394:485-490.

Cassella JP, Lawrenson JG and Firth JA (1997): Development of endothelial paracellular clefts and their tight junctions in the pial microvessels of the rat. J Neurocytol 26:567-575.

Cao J, Schulte J, Knight A, Leslie NR, Zagozdzon A, Bronson R, Manevich Y, Beeson C and Neumann CA (2009): Prdx1 inhibits tumorigenesis via regulating PTEN/AKT activity. EMBO J 28:1505-1517.

Central Brain Tumor Registry of the United States (2006)

Cha MK, Suh KH and Kim IH (2009): Overexpression of peroxiredoxin I and thioredoxin1 in human breast carcinoma. J Exp Clin Cancer Res 28:93.

Chang TS, Jeong W, Woo HA, Lee SM, Park S and Rhee SG (2004): Characterization of mammalian sulfiredoxin and its reactivation of hyperoxidized peroxiredoxin through reduction of cysteine sulfinic acid in the active site to cysteine. J Biol Chem 279:50994-51001. 74

Chang XZ, Li DQ, Hou YF, Wu J, Lu JS, Di GH, Jin W, Ou ZL, Shen ZZ and Shao ZM (2007): Identification of the functional role of peroxiredoxin 6 in the progression of breast cancer. Breast Cancer Res 9:R76.

Cheng Y, Pang JC, Ng HK, Ding M, Zhang SF, Zheng J, Liu DG and Poon WS (2000): Pilocytic astrocytomas do not show most of the genetic changes cmmonly seen in diffuse astrocytomas. Histopathology 37:437-444.

Cheung ST, Leung KL, Ip YC, Chen X, Fong DY, Ng IO, Fan ST and So S (2005): Claudin-10 expression level is associated with recurrence of primary hepatocellular carcinoma. Clin Cancer Res 11:551-556.

Chia SK, Wykoff CC, Watson PH, Han C, Leek RD, Pastorek J, Gatter KC, Ratcliffe P and Harris AL (2001): Prognostic significance of a novel hypoxia-regulated marker, carbonic anhydrase IX, in invasive breast carcinoma. J Clin Oncol 19:3660-3668.

Chiche J, Ilc K, Laferrière J, Trottier E, Dayan F, Mazure NM, Brahimi-Horn MC and Pouysségur J (2009): Hypoxia-inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH. Cancer Res 69:358-368.

Choi JH, Kim TN, Kim S, Baek S-H, Kim J-H, Lee SR, Kim JR (2002): Overexpression of mitochondrial thioredoxin reductase and peroxiredoxin iii in hepatocellular carcinomas. Anticancer Res 22:3331-3336.

Chrastina A, Závada J, Parkkila S, Kaluz S, Kaluzová M, Rajcáni J, Pastorek J and Pastoreková S (2003): Biodistribution and pharmacokinetics of 125I-labeled monoclonal antibody M75 specific for carbonic anhydrase IX, an intrinsic marker of hypoxia, in nude mice xenografted with human colorectal carcinoma. Int J Cancer 105:873-881.

Christianson DW and Cox JD (1999): Catalysis by metal-activated hydroxide in zinc and manganese metalloenzymes. Annu Rev Biochem 68:33-57.

75

Chua PJ, Lee EH, Yu Y, Yip GW, Tan PH and Bay BH (2010): Silencing the Peroxiredoxin III gene inhibits cell proliferation in breast cancer. Int J Oncol 36:359-364.

Chung YM, Yoo YD, Park JK, Kim YT and Kim HJ (2001): Increased expression of peroxiredoxin II confers resistance to cisplatin. Anticancer Res 21:1129-1133.

Church SL, Grant JW, Meese EU and Trent JM (1992): Sublocalization of the gene encoding manganese superoxide dismutase (MnSOD/SOD2) to 6q25 by fluorescence in situ hybridization and somatic cell hybrid mapping. Genomics 14:823-825.

Cohen TS, Gray Lawrence G and Margulies SS (2010): Cultured alveolar epithelial cells from septic rats mimic in vivo septic lung. PLoS One 5:e11322.

Combs SE, Thilmann C, Debus J and Schulz-Ertner D (2006): Local radiotherapeutic management of ependymomas with fractionated stereotactic radiotherapy (FSRT). BMC Cancer 6:222.

Coyne CB, Gambling TM, Boucher RC, Carson JL and Johnson LG (2003): Role of claudin interactions in airway tight junctional permeability. Am J Physiol Lung Cell Mol Physiol 285:L1166-1178.

Dalton TP, Dieter MZ, Yang Y, Shertzer HG and Nebert DW (2000): Knockout of the mouse glutamate cysteine ligase catalytic subunit (Gclc) gene: embryonic lethal when homozygous, and proposed model for moderate glutathione deficiency when heterozygous. Biochem Biophys Res Commun 279:324-329.

Das B, Yeger H, Tsuchida R, Torkin R, Gee MF, Thorner PS, Shibuya M, Malkin D and Baruchel S (2005): A hypoxia-driven vascular endothelial growth factor/Flt1 autocrine loop interacts with hypoxia-inducible factor-1alpha through mitogen-activated protein kinase/extracellular signalregulated kinase 1/2 pathway in neuroblastoma. Cancer Res 65:7267-7275.

Daugherty BL, Mateescu M, Patel AS, Wade K, Kimura S, Gonzales LW, Guttentag S, Ballard PL and Koval M (2004): Developmental regulation of claudin localization by fetal alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 287: L1266-1273.

76

Demasi AP, Costa AF, Altemani A, Furuse C, Araújo NS and Araújo VC (2010): Glucose transporter protein 1 expression in mucoepidermoid carcinoma of salivary gland: correlation with grade of malignancy. Int J Exp Pathol 91:107-113.

Deutsch M, Thomas PR, Krischer J, Boyett JM, Albright L, Aronin P, Langston J, Allen JC, Packer RJ, Linggood R, Mulhern R, Stanley P, Stehbens JA, Duffner P, Kun L, Rorke L, Cherlow J, Freidman H, Finlay JL and Vietti T (1996): Results of a prospective randomised trial comparing standard dose neuraxis irradiation (3,600 cGy/20) with reduced neuraxis irradiation (2,340 cGy/13) in patients with low-stage medulloblastoma. A Combined Children´s Cancer Group-Pediatric Oncology Group study. Pediatr Neurosurg 24:167-176.

Dirks PB, Jay V, Becker LE, Drake JM, Humphreys RP, Hoffman HJ and Rutka JT (1994): Development of anaplastic changes in low-grade astrocytomas of childhood. Neurosurgery 34:6878.

D'Souza T, Agarwal R and Morin PJ (2005): Phosphorylation of claudin-3 at threonine 192 by cAMP-dependent protein kinase regulates tight junction barrier function in ovarian cancer cells. J Biol Chem 280:26233-26240.

Duncan JA and Hoffman HJ (1995): Intracranial ependymomas. In: Brain Tumors. Kaye AH, Lows ER, Jr., eds. Churchill Livingstone: Edinburgh, pp.493-504

Eberhart CG, Kepner JL, Goldthwaite PT, Kun LE, Duffner PK, Friedman HS, Strother DR and Burger BC (2002): Histopathologic grading of medulloblastomas: a Pediatric Oncology Group study. Cancer 94:552-560.

Eismann T, Huber N, Shin T, Kuboki S, Galloway E, Wyder M, Edwards MJ, Greis KD, Shertzer HG, Fisher AB and Lentsch AB (2009): Peroxiredoxin-6 protects against mitochondrial dysfunction and liver injury during ischemia-reperfusion in mice. Am J Physiol Gastrointest Liver Physiol 296:G266-274.

Ellison DW (2010): Childhood medulloblastoma: novel approaches to the classification of a heterogenous disease. Acta Neuropathologica 120:305-316.

77

Ellison DW, Clifford SC, Gajjar A and Gilbertson RJ (2003): What´s new in neuro-oncology? Recent advances in medulloblastoma. Eur J Paediatr Neurol 7:53-66.

Enoksson M, Fernandes AP, Prast S, Lillig CH, Holmgren A and Orrenius S (2005): Overexpression of glutaredoxin 2 attenuates apoptosis by preventing cytochrome c release. Biochem Biophys Res Commun 327:774-779.

Ernestus RI, Schröder R, Stützer H and Klug N (1996): Prognostic relevance of localization and grading in intracranial ependymomas of childhood. Childs Nerv Syst 12:522-526.

Evans AE, Jenkin RD, Sposto R, Ortega JA, Wilson CB, Wara W, Ertel IJ, Kramer S, Chang CH, Leikin SL and Hammond GD (1990): The treatment of medulloblastoma. Results of a prospective randomised trial of radiation therapy with and without CCNU, vincristine, and prednisone. J Neurosurg 72:572-582.

Evans AE, Anderson JR, Lefkowitz-Boudreaux IB, and Finlay JL (1996): Adjuvant chemotherapy of childhood posterior fossa ependymoma: cranio-spinal irradiation with or without adjuvant CCNU, vincristine, and prednisone: a Childrens Cancer Group study. Med Pediatr Oncol 27:8-14.

Fangusaro J, Finlay J, Sposto R, Ji L, Saly M, Zacharoulis S, Asgharzadeh S, Abromowitch M, Olshefski R, Halpern S, Dubowy R, Comito M, Diez B, Kellie S, Hukin J, Rosenblum M, Dunkel I, Miller DC, Allen J and Gardner S (2008): Intensive chemotherapy followed by consolidative myeloablative chemotherapy with autologous hematopoietic cell rescue (AuHCR) in young children with newly diagnosed supratentorial primitive neuroectodermal tumors (sPNETs): report of the Head Start I and II experience. Pediatr Blood Cancer 50:312-318.

Farina AR, Tacconelli A, Cappabianca L, Masciulli MP, Holmgren A, Beckett GJ, Gulino A and Mackay AR (2001): Thioredoxin alters the matrix metalloproteinase/tissue inhibitors of metallopriteinase balance and stimulates human SK-N-SH neuroblastoma cell invasion. Eur J Biochem 268:405-413.

Fatma N, Singh DP, Shinohara T and Chylack LT Jr (2001): Transcriptional regulation of the antioxidant protein 2 gene, a thiol-specific antioxidant, by lens epithelium-derived growth factor to protect cells from oxidative stress. J Biol Chem 276:48899-48907. 78

Fernandez C, Figarella-Branger D, Girard N, Bouvier-Labit C, Gouvernet J, Paz Paredes A and Lena G (2003): Pilocytic astrocytomas in children: prognostic factors--a retrospective study of 80 cases. Neurosurgery 53:544-553.

Fischer B, Müller B, Fischer KG, Baur N and Kreutz W (2000): Acidic pH inhibits non-MHCrestricted killer cell functions. Clin Immunol 96:252-263.

Frank S, Munz B and Werner S (1997): The human homologue of a bovine non-selenium glutathione peroxidase is a novel keratinocyte growth factor-regulated gene. Oncogene 14:915-921.

Fujii T, Fujii J and Taniguchi N (2001): Augmented expression of peroxiredoxin VI in rat lung and kidney after birth implies an antioxidative role. Eur J Biochem 268:218-225.

Fukumura D, Xu L, Chen Y, Gohongi T, Seed B and Jain RK (2001): Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Res 61:6020-6024.

Fulham MJ, Melisi JW, Nishimiya J, Dwyer AJ and Di Chiro G (1993): Neuroimaging of juvenile pilocytic astrocytomas: an enigma. Radiology 189:221-225.

Furie DM and Provenzale JM (1995): Supratentorial ependymomas and subependymomas: CT and MR appearance. J Comput Assist Tomogr 19:518-526.

Furuse M, Hirase T, Itoh M, Nagafuchi A, Yonemura S, Tsukita S, Tsukita S (1993): Occludin: a novel integral membrane protein localizing at tight junctions. J Cell Biol 123:1777-1788.

Furuse M, Sasaki, H. Fujimoto, S and Tsukita A (1998): single gene product, claudin-1 or -2, reconstitutes tight junction strands and recruits occludin in fibroblasts. J. Cell Biol 143:391-401.

Gan L, Yang XL, Liu Q and Xu HB (2005): Inhibitory effects of thioredoxin reductase antisense RNA on the growth of human hepatocellular carcinoma cells. J Cell Biochem 96:653-664.

79

Gatta G, Capocaccia R, Stiller C, Kaatsch P, Berrino F and Terenziani M; EUROCARE Working Group (2005): Childhood cancer survival trends in Europe: a EUROCARE Working Group study. J Clin Oncol 23:3742-3751.

Gerweck LE (1998): Tumor pH: implications for treatment and novel drug design. Semin Radiat Oncol 8:176-182.

Geyer JR, Zeltzer PM, Boyett JM, Rorke LB, Stanley P, Albright AL, Wisoff JH, Milstein JM, Allen JC, Finlay JL, Ayers GD, Shurin SB, Stevens KR and Bleyer WA (1994): Survival of infants with primitive neurectodermal tumors or malignant ependymomas of the CNS treated with eight drugs in 1 day: a report from the Children´s Cancer Group 12:1607-1615.

Geyer JR, Sposto R, Jennings M, Boyett JM, Axtell RA, Breiger D, Broxon E, Donahue B, Finlay JL, Goldwein JW, Heier LA, Johnson D, Mazewski C, Miller DC, Packer R, Puccetti D, Radcliffe J, Tao ML and Shiminski-Maher T (2005): Multiagent chemotherapy and deferred radiotherapy in infants with malignant brain tumors: a report from the Children´s Cancer Group. J Clin Oncol 23:7621-7631.

Ghosh J and Myers CE (1998): Inhibition of arachidonate 5-lipoxygenase triggers massive apoptosis in human prostate cancer cells. Proc Natl Acad Sci U S A 95:13182-13187.

Giangaspero F, Perilongo G, Fondelli MP, Brisigotti M, Carollo C, Burnelli R, Burger PC and Garre ML (1999): Medulloblastoma with extensive nodularity: a variant with favorable outcome. J Neurosurg 91:971-977.

Grabmaier K, Vissers JL, De Weijert MC, Oosterwijk-Wakka JC, Van Bokhoven A, Brakenhoff RH, Noessner E, Mulders PA, Merkx G, Figdor CG, Adema GJ and Oosterwijk E (2000): Molecular cloning and immunogenicity of renal cell carcinoma-associated antigen G250. Int J Cancer 85:865-870.

Grebenkämper K and Galla HJ (1994): Translational diffusion measurements of a fluorescent phospholipid between MDCK-1 cells support the lipid model of the tight junctions. Chem. Phys. Lipids 71:133-143.

80

Greiner J, Schmitt M, Li L, Giannopoulos K, Bosch K, Schmitt A, Dohner K, Schlenk RF, Pollack JR, Dohner H and Bullinger L (2006): Expression of tumor-associated antigens in acute myeloid leukemia: Implications for specific immunotherapeutic approaches. Blood 108:4109-4117.

Griffith OW (1999): Biologic and pharmacologic regulation of mammalian glutathione synthesis. Free Radic. Biol. Med 27:922–935.

Griffith OW and Mulcahy RT (1999): The enzymes of glutathione synthesis: gammaglutamylcysteine synthetase. Adv. Enzymol. Relat. Areas Mol. Biol 73:209–267.

Guyton KZ and Kensler TW (1993): Oxidative mechanisms in carcinogenesis. Br Med Bull 49:523544.

Haapasalo H, Sallinen S, Sallinen P, Helén P, Jääskeläinen J, Salmi TT, Paetau A, Paljärvi L, Visakorpi T and Kalimo H (1999): Clinicopathological correlation of cell proliferation, apoptosis and p53 in cerebellar pilocytic astrocytomas. Neuropathol Appl Neurobiol 25:134-142.

Haapasalo H, Kyläniemi M, Paunul N, Kinnula VL and Soini Y (2003): Expression of antioxidant enzymes in astrocytic brain tumors. Brain Pathol 13:155-164.

Haapasalo JA, Nordfors KM, Hilvo M, Rantala IJ, Soini Y, Parkkila AK, Pastoreková S, Pastorek J, Parkkila SM and Haapasalo HK (2006): Expression of carbonic anhydrase IX in astrocytic tumors predicts poor prognosis. Clin Cancer Res 12:473-477.

Haapasalo J, Nordfors K, Järvelä S, Bragge H, Rantala I, Parkkila AK, Haapasalo H and Parkkila S (2007): Carbonic anhydrase II in the endothelium of glial tumors: a potential target for therapy. Neuro Oncol 9:308-313.

Haapasalo J, Hilvo M, Nordfors K, Haapasalo H, Parkkila S, Hyrskyluoto A, Rantala I, Waheed A, Sly WS, Pastorekova S, Pastorek J and Parkkila AK (2008): Identification of an alternatively spliced isoform of carbonic anhydrase XII in diffusely infiltrating astrocytic gliomas. Neuro Oncol 10:131-138.

81

Halliwell B (1991): Reactive oxygen species in living systems: source, biochemistry, and role in human disease. Am J Med 91:14S-22S.

Halliwell B (2007): Oxidative stress and cancer: have we moved forward? Biochem J 401:1-11.

Halliwell B and Gutteridge JMC (2007): Free Radicals in Biology and Medicine. New York: Oxford University Press.

Han H, Bearss DJ, Browne LW, Calaluce R, Nagle RB and Von Hoff DD (2002): Identification of differentially expressed genes in pancreatic cancer cells using cdna microarrays. Cancer Res 62:2890-2896.

Hansen JM, Moriarty-Craige S and Jones DP (2007): Nuclear and cytoplasmic peroxiredoxin-1 differentially regulate NF-kappaB activities. Free Radic Biol Med 43:282-288.

Hattori F, Murayama N, Noshita T and Oikawa S (2003): Mitochondrial peroxiredoxin-3 protects hippocampal neurons from excitotoxic injury in vivo. J Neurochem 86:860-868.

Hedley D, Pintilie M, Woo J, Nicklee T, Morrison A, Birle D, Fyles A, Milosevic M and Hill R (2004): Up-regulation of the redox mediators thioredoxin and apurinic/apyrimidinic excision (APE)/Ref-1 in hypoxic microregions of invasive cervical carcinomas, mapped using multispectral, wide-field fluorescence image analysis. Am J Pathol 164:557-565.

Hewett-Emmett D and Tashian RE (1996): Functional diversity, conservation, and convergence in the evolution of the alpha-, beta-, and gamma-carbonic anhydrase gene families. Mol Phylogenet Evol 5:50-77.

Hewitt KJ, Agarwal R and Morin PJ (2006): The claudin gene family: expression in normal and neoplastic tissues. BMC Cancer 6:186.

Hilvo M, Baranauskiene L, Salzano AM, Scaloni A, Matulis D, Innocenti A, Scozzafava A, Monti SM, Di Fiore A, De Simone G, Lindfors M, Jänis J, Valjakka J, Pastoreková S, Pastorek J, Kulomaa MS, Nordlund HR, Supuran CT and Parkkila S (2008): Biochemical characterization of CA IX, one of the most active carbonic anhydrase isozymes. J Biol Chem 283:27799-27809. 82

Holmes JL, Van Itallie CM, Rasmussen JE and Anderson JM (2006): Claudin profiling in the mouse during postnatal intestinal development and along the gastrointestinal tract reveals complex expression patterns. Gene Expr Patterns 6:581-588.

Horn B, Heideman R, Geyer R, Pollack I, Packer R, Goldwein J, Tomita T, Schomberg P, Ater J, Luchtman-Jones L, Rivlin K, Lamborn K, Prados M, Bollen A, Berger M, Dahl G, McNeil E, Patterson K, Shaw D, Kubalik M and Russo C (1999): A multi-institutional retrospective study of intracranial ependymoma in children: identification of risk factors. J Pediatr Hematol Oncol 21:203211.

Horten BC and Rubinstein LJ (1976): Primary cerebral neuroblastoma. A clinicopathological study of 35 cases. Brain 99:735-756.

Hoskin PJ, Sibtain A, Daley FM and Wilson GD (2003): GLUT1 and CAIX as intrinsic markers of hypoxia in bladder cancer: relationship with vascularity and proliferation as predictors of outcome of ARCON. J Cancer 89:1290-1297.

Hough CD, Sherman-Baust CA, Pizer ES, Montz FJ, Im DD, Rosenshein NB, Cho KR, Riggins GJ and Morin PJ (2000): Large-scale serial analysis of gene expression reveals genes differentially expressed in ovarian cancer. Cancer Res 60:6281-6287.

Hoyt WF and Baghdassarian SA (1969): Optic glioma of childhood. Natural history and rationale for conservative management. Br J Ophthalmol 53:793-798.

Hussein D, Estlin EJ, Dive C and Makin GW (2006): Chronic hypoxia promotes hypoxia-inducible factor-1alpha-dependent resistance to etoposide and vincristine in neuroblastoma cells. Mol Cancer Ther 5:2241-2250.

Ilveskoski I, Pihko H, Wiklund T, Lamminranta S, Perkkiö M, Mäkipernaa A, Salmi TT, Lanning M and Saarinen UM (1996): Neuropsychologic late effects in children with malignant brain tumors treated with surgery, radiotherapy and "8 in 1" chemotherapy. Neuropediatrics 27:124-129.

International Human Genome Sequencing Consortium (2001): Nature 409:860-921.

83

Ishii T, Itoh K, Takahashi S, Sato H, Yanagawa T, Katoh Y, Bannai S, and Yamamoto M (2000): Transcription factor Nrf2 coordinately regulates a group of oxidative stress-inducible genes in macrophages. J Biol Chem 275:16023-16029.

Ishikawa T, Bao JJ, Yamane Y, Akimaru K, Frindrich K, Wright CD and Kuo MT (1996): Coordinated induction of MRP ⁄ GS-X pump and gamma-glutamylcysteine synthetase by heavy metals in human leukemia cells. J. Biol. Chem 271:14981–14988.

Ishizaki T, Chiba H, Kojima T, Fujibe M, Soma T, Miyajima H, Nagasawa K, Wada I and Sawada N (2003): Cyclic AMP induces phosphorylation of claudin-5 immunoprecipitates and expression of claudin-5 gene in blood-brain-barrier endothelial cells via protein kinase A-dependent and independent pathways. Exp Cell Res 290:275-288.

Ivanov SV, Kuzmin I, Wei MH, Pack S, Geil L, Johnson BE, Stanbridge EJ and Lerman MI (1998): Down-regulation of transmembrane carbonic anhydrases in renal cell carcinoma cell lines by wildtype von Hippel-Lindau transgenes. Proc Natl Acad Sci U S A 95:12596-12601.

Ivanov S, Liao SY, Ivanova A, Danilkovitch-Miagkova A, Tarasova N, Weirich G, Merrill MJ, Proescholdt MA, Oldfield EH, Lee J, Zavada J, Waheed A, Sly W, Lerman MI and Stanbridge EJ (2001): Expression of hypoxia-inducible cell-surface transmembrane carbonic anhydrases in human cancer. Am J Pathol 158:905-919.

Jakacki RI (2005): Treatment strategies for high-risk medulloblastoma and supratentorial primitive neuroectodermal tumors: Rewiev of the literature. J Neurosurg 102:44-52.

Jayawickreme DP, Hayward RD and Harkness WF (1995): Intracranial ependymomas in childhood: a report of 24 cases followed for 5 years. Childs Nerv Syst 11:409-413.

Jikimoto T, Nishikubo Y, Koshiba M, Kanagawa S, Morinobu S, Morinobu A, Saura R, Mizuno K, Kondo S, Toyokuni S, Nakamura H, Yodoi J and Kumagai S (2002): Thioredoxin as a biomarker for oxidative stress in patients with rheumatoid arthritis. Mol Immunol 38:765-772.

Jin DY, Chae HZ, Rhee SG and Jeang KT (1997): Regulatory role for a novel human thioredoxin peroxidase in NF-kappaB activation. J Biol Chem 272:30952-30961. 84

Jin MH, Lee YH, Kim JM, Sun HN, Moon EY, Shong MH, Kim SU, Lee SH, Lee TH, Yu DY and Lee DS (2005): Characterization of neural cell types expressing peroxiredoxins in mouse brain. Neurosci Lett 381:252-257.

Johanson C (1980): Permeability and vascularity of the developing brain: Cerebellum vs. cerebral cortex. Brain Res 190:3-16.

Johnston DL, Keene DL, Lafay-Cousin L, Steinbok P, Sung L, Carret AS, Crooks B, Strother D, Wilson B, Odame I, Eisenstat DD, Mpofu C, Zelcer S, Huang A and Bouffet E (2008): Supratentorial primitive neuroectodermal tumors: a Canadian pediatric brain tumor consortium report. J Neurooncol 86:101-108.

Jones DT, Ichimura K, Liu L, Pearson DM, Plant K and Collins VP (2006): Genomic analysis of pilocytic astrocytomas at 0.97 Mb resolution shows an increasing tendency toward chromosomal copy number change with age. J Neuropathol Exp Neurol 65:1049-1058.

Jornot L, Petersen H and Junod AF (1998): Hydrogen peroxide-induced DNA damage is independent of nuclear calcium but dependent on redox-active ions. Biochem J 335:85-94.

Järvelä S, Bragge H, Paunu N, Järvelä T, Paljärvi L, Kalimo H, Helén P, Kinnula V, Soini Y and Haapasalo H (2006): Antioxidant enzymes in oligodendroglial brain tumors: association with proliferation, apoptotic activity and survival. J Neurooncol 77:131-140.

Järvelä S, Rantala I, Rodriguez A, Kallio H, Parkkila S, Kinnula VL, Soini Y and Haapasalo H (2010): Specific expression profile and prognostic significance of peroxiredoxins in grade II-IV astrocytic brain tumors. BMC Cancer 10:104.

Järvinen K, Pietarinen-Runtti P, Linnainmaa K, Raivio KO, Krejsa CM, Kavanagh T and Kinnula VL (2000): Antioxidant defense mechanisms of human mesothelioma and lung adenocarcinoma cells. Am. J. Physiol. Lung Cell Mol. Physiol 278:L696–702.

85

Kaarteenaho-Wiik R and Soini Y (2009): Claudin-1, -2, -3, -4, -5, and -7 in usual interstitial pneumonia and sarcoidosis. J Histochem Cytochem 57:187-195.

Kaarteenaho R, Merikallio H, Lehtonen S, Harju T and Soini Y (2010): Divergent expression of claudin -1, -3, -4, -5 and -7 in developing human lung. Respir Res 11:59.

Kagawa N, Maruno M, Suzuki T, Hashiba T, Hashimoto N, Izumoto S and Yoshimine T (2006): Detection of genetic and chromosomal aberrations in medulloblastomas and primitive neuroectodermal tumors with DNA microarrays. Brain Tumor Pathol 23:41-47.

Kahlos K, Pääkkö P, Kurttila E, Soini Y and Kinnula VL (2000): Manganese superoxide dismutase as a diagnostic marker for malignant pleural mesothelioma. Br J Cancer 82:1022-1029.

Kallio H, Pastorekova S, Pastorek J, Waheed A, Sly WS, Mannisto S, Heikinheimo M and Parkkila S (2006): Expression of carbonic anhydrases IX and XII during mouse embryonic development. BMC Dev Biol 6:22.

Kang SW, Baines IC and Rhee SG (1998a): Characterization of a mammalian peroxiredoxin that contains one conserved cysteine. J Biol Chem 273:6303-6311.

Kang SW, Chae HZ, Seo MS, Kim K, Baines IC and Rhee SG (1998b): Mammalian peroxiredoxin isoforms can reduce hydrogen peroxide generated in response to growth factors and tumor necrosis factor-alpha. J Biol Chem 273:6297-6302.

Karhumaa P, Parkkila S, Türeci O, Waheed A, Grubb JH, Shah G, Parkkila A, Kaunisto K, Tapanainen J, Sly WS and Rajaniemi H (2000): Identification of carbonic anhydrase XII as the membrane isozyme expressed in the normal human endometrial epithelium. Mol Hum Reprod 6:6874.

Karhumaa P, Kaunisto K, Parkkila S, Waheed A, Pastoreková S, Pastorek J, Sly WS and Rajaniemi H (2001): Expression of the transmembrane carbonic anhydrases, CA IX and CA XII, in the human male excurrent ducts. Mol Hum Reprod 7:611-616.

86

Karihtala P, Mäntyniemi A, Kang SW, Kinnula VL and Soini Y (2003): Peroxiredoxins in breast carcinoma. Clin Cancer Res 9:3418-3424.

Kato Y, Nakayama Y, Umeda M and Miyazaki K (1992): Induction of 103-kDa gelatinase/type IV collagenase by acidic culture conditions in mouse metastatic melanoma cell lines. J Biol Chem 267:11424-11430.

Kida E, Palminiello S, Golabek AA, Walus M, Wierzba-Bobrowicz T, Rabe A, Albertini G and Wisniewski KE (2006): Carbonic anhydrase II in the developing and adult human brain. J Neuropathol Exp Neurol 65:664-674.

Kieran MW, Walker D, Frappaz D, Prados M (2010): Brain tumors: from childhood through adolescence into adulthood. J Clin Oncol 28:4783-4789.

Kim HJ, Chae HZ, Kim YJ, Kim YH, Hwang TS, Park EM and Park YM (2003): Preferential elevation of prx i and trx expression in lung cancer cells following hypoxia and in human lung cancer tissues. Cell Biol Toxicol 19:285-298.

Kim HS, Pak JH, Gonzales LW, Feinstein SI and Fisher AB (2002): Regulation of cys-1 peroxiredoxin expression in lung epithelial cells. Am J. Respir. Cell Mol. Biol 27:227-233.

Kim JH, Bogner PN, Ramnath N, Park Y, Yu J and Park YM (2007): Elevated peroxiredoxin 1, but not NF-E2-related factor 2, is an independent prognostic factor for disease recurrence and reduced survival in stage I non-small cell lung cancer. Clin Cancer Res 13:3875-3882.

Kim SH, Fountoulakis M, Cairns N and Lubec G (2001): Protein levels of human peroxiredoxin subtypes in brains of patients with Alzheimer's disease and Down syndrome. J Neural Transm Suppl 223-235.

Kim SU, Hwang CN, Sun HN, Jin MH, Han YH, Lee H, Kim JM, Kim SK, Yu DY, Lee DS and Lee SH (2008): Peroxiredoxin I is an indicator of microglia activation and protects against hydrogen peroxide-mediated microglial death. Biol Pharm Bull 31:820-825.

87

Kinnula VL and Crapo JD (2004): Superoxide dismutases in malignant cells and human tumors. Free Radic Biol Med 36:718-744.

Kinnula VL, Crapo JD and Raivio KO (1995): Generation and disposal of reactive oxygen metabolites in the lung. Lab Invest 73:3-19.

Kinnula VL, Lehtonen S, Sormunen R, Kaarteenaho-Wiik R, Kang SW, Rhee SG and Soini Y (2002): Overexpression of peroxiredoxins I, II, III, V, and VI in malignant mesothelioma. J Pathol 196:316-323.

Kino-Ohsaki J, Nishimori I, Morita M, Okazaki K, Yamamoto Y, Onishi S and Hollingsworth MA (1996): Serum antibodies to carbonic anhydrase I and II in patients with idiopathic chronic pancreatitis and Sjögren's syndrome. Gastroenterology 110:1579-1586.

Kinscherf R, Claus R, Wagner M, Gehrke C, Kamencic H, Hou D, Nauen O, Schmiedt W, Kovacs G, Pill J, Metz J and Deigner HP (1998): Apoptosis caused by oxidized LDL is manganese superoxide dismutase and p53 dependent. FASEB J 12:461-467.

Kiuchi-Saishin Y, Gotoh S, Furuse M, Takasuga A, Tano Y and Tsukita S (2002): Differential expression patterns of claudins, tight junction membrane proteins, in mouse nephron segments. J Am Soc Nephrol 13:875-886.

Kivelä AJ, Parkkila S, Saarnio J, Karttunen TJ, Kivelä J, Parkkila AK, Pastoreková S, Pastorek J, Waheed A, Sly WS and Rajaniemi H (2000a): Expression of transmembrane carbonic anhydrase isoenzymes IX and XII in normal human pancreas and pancreatic tumours. Histochem Cell Biol 114:197-204.

Kivelä A, Parkkila S, Saarnio J, Karttunen TJ, Kivelä J, Parkkila AK, Waheed A, Sly WS, Grubb JH, Shah G, Türeci O and Rajaniemi H (2000b): Expression of a novel transmembrane carbonic anhydrase isozyme XII in normal human gut and colorectal tumors. Am J Pathol 56:577-584.

Kniesel U and Wolburg H (2000): Tight Junctions of the Blood-Brain Barrier. Cell Mol Neurobiol 20:57-76.

88

Knoops B, Clippe A, Bogard C, Arsalane K, Wattiez R, Hermans C, Duconseille E, Falmagne P and Bernard A (1999): Cloning and characterization of AOEB166, a novel mammalian antioxidant enzyme of the peroxiredoxin family. J Biol Chem 274:30451-30458.

Koda R, Zhao L, Yaoita E, Yoshida Y, Tsukita S, Tamura A, Nameta M, Zhang Y, Fujinaka H, Magdeldin S, Xu B, Narita I and Yamamoto T (2011): Novel expression of claudin-5 in glomerular podocytes. Cell Tissue Res 343:637-648.

Kominsky SL, Argani P, Korz D, Evron E, Raman V, Garrett E, Rein A, Sauter G, Kallioniemi OP and Sukumar S (2003): Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal carcinoma of the breast. Oncogene 22:2021-2033.

Kominsky SL, Vali M, Korz D, Gabig TG, Weitzman SA, Argani P and Sukumar S (2004): Clostridium perfringens enterotoxin elicits rapid and specific cytolysis of breast carcinoma cells mediated through tight junction proteins claudin 3 and 4. Am J Pathol 164:1627-1633.

Korshunov A, Golanov A, Sycheva R and 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.

Koto T, Takubo K, Ishida S, Shinoda H, Inoue M, Tsubota K, Okada Y and Ikeda E (2007): Hypoxia disrupts the barrier function of neural blood vessels through changes in the expression of claudin-5 in endothelial cells. Am J Pathol 170:1389–1397.

Koukourakis MI, Giatromanolaki A, Sivridis E, Simopoulos K, Pastorek J, Wykoff CC, Gatter KC and Harris AL (2001): Hypoxia-regulated carbonic anhydrase-9 (CA9) relates to poor vascularization and resistance of squamous cell head and neck cancer to chemoradiotherapy. Clin Cancer Res 7:3399-3403.

Krasnapolski MA, Todaro LB and De Kier Joffe EB (2011): Is the Epithelial-to-Mesenchymal Transition Clinically Relevant for the Cancer Patient? Curr Pharm Biotechnol Apr 6. [Epub ahead of print].

89

Krause G, Winkler R, Mueller SL, Haseloff RL, Piontek J and Blasig IE (2008): Structure and function of claudins. Biochimica et Biophysica Acta 1778:631-645.

Krämer F, White K, Kubbies M, Swisshelm K and Weber BH (2000): Genomic organization of claudin-1 and its assessment in hereditary and sporadic breast cancer. Hum Genet 107:249-256.

Kumpulainen T, Dahl D, Korhonen LK and Nyström SH (1983): Immunolabeling of carbonic anhydrase isoenzyme C and glial fibrillary acidic protein in paraffin-embedded tissue sections of human brain and retina. J Histochem Cytochem 31:879-886.

Kurt E, Zheng PP, Hop WC, van der Weiden M, Bol M, van den Bent MJ, Avezaat CJ and Kros JM (2006): Identification of relevant prognostic histopathologic features in 69 intracranial ependymomas, excluding myxopapillary ependymomas and subependymomas. Cancer 106:388395.

Lannering B, Marky I, Lundberg A and Ohlsson E (1990): Long-term sequelae after pediatric brain tumors – their effect on disability and quality of life. Med Ped Oncol 18:304-310.

Laws ER, Taylor WF, Clifton MB and Okazaki H (1984): Neurosurgical management of low-grade astrocytoma of the cerebral hemispheres. J. Neurosurg 61:665-673.

Lechpammer M, Resnick MB, Sabo E, Yakirevich E, Greaves WO, Sciandra KT, Tavares R, Noble LC, DeLellis RA and Wang LJ (2008): The diagnostic and prognostic utility of claudin expression in renal cell neoplasms. Mod Pathol 21:1320-1329.

Lee SB, Ho JN, Yoon SH, Kang GY, Hwang SG and Um HD (2009): Peroxiredoxin 6 promotes lung cancer cell invasion by inducing urokinase-type plasminogen activator via p38 kinase, phosphoinositide 3-kinase, and Akt. Mol Cells 28:583-588.

Lee SK, Moon J, Park SW, Song SY, Chung JB and Kang JK (2005): Loss of the tight junction protein claudin 4 correlates with histological growth-pattern and differentiation in advanced gastric adenocarcinoma. Oncol Rep 13:193-199.

90

Lee TH, Kim SU, Yu SL, Kim SH, Park DS, Moon HB, Dho SH, Kwon KS, Kwon HJ, Han YH, Jeong S, Kang SW, Shin HS, Lee KK, Rhee SG and Yu DY (2003): Peroxiredoxin II is essential for sustaining life span of erythrocytes in mice. Blood 101:5033-5038.

Lee YJ, Galoforo SS, Berns CM, Chen JC, Davis BH, Sim JE, Corry PM and Spitz DR (1998): Glucose deprivation-induced cytotoxicity and alterations in mitogen-activated protein kinase activation are mediated by oxidative stress in multidrug-resistant human breast carcinoma cells. J Biol Chem 273:5294-5299.

Lee YY, Van Tassel P, Bruner JM, Moser RP and Share JC (1989): Juvenile pilocytic astrocytomas: CT and MR characteristics. AJR Am J Roentgenol 152:1263-1270.

Leppilampi M, Koistinen P, Savolainen ER, Hannuksela J, Parkkila AK, Niemelä O, Pastoreková S, Pastorek J, Waheed A, Sly WS, Parkkila S and Rajaniemi H (2002): The expression of carbonic anhydrase II in hematological malignancies. Clin Cancer Res 8:2240-2245.

Leppilampi M, Saarnio J, Karttunen TJ, Kivelä J, Pastoreková S, Pastorek J, Waheed A, Sly WS and Parkkila S (2003): Carbonic anhydrase isozymes IX and XII in gastric tumors. World J Gastroenterol 9:1398-1403.

Liao SY, Brewer C, Závada J, Pastorek J, Pastorekova S, Manetta A, Berman ML, DiSaia PJ and Stanbridge EJ (1994): Identification of the MN antigen as a diagnostic biomarker of cervical intraepithelial squamous and glandular neoplasia and cervical carcinomas. Am J Pathol 145:598609.

Liao SY, Aurelio ON, Jan K, Zavada J and Stanbridge EJ (1997): Identification of the MN/CA9 protein as a reliable diagnostic biomarker of clear cell carcinoma of the kidney. Cancer Res 57:2827-2831.

Liebner S, Fischmann A, Rascher G, Duffner F, Grote EH, Kalbacher H and Wolburg H (2000): Claudin-1 and claudin-5 expression and tight junction morphology are altered in blood vessels of human glioblastoma multiforme. Acta Neuropathol 100:323-331.

91

Li D and Mrsny RJ (2000): Oncogenic Raf-1 disrupts epithelial tight junctions via downregulation of occludin. J Cell Biol 148:791-800.

Li JJ, Oberley L, St Clair DK, Ridnour LA and Oberley TD (1995a): Phenotypic changes induced in human breast cancer cells by overexpression of manganese-containing superoxide dismutase. Oncogene 10:1989-2000.

Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, Berger C, Chan PH, Wallace DC and Epstein CJ (1995b): Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet 11:376-381.

Lim JC, Choi HI, Park YS Nam HW, Woo HA, Kwon KS, Kim YS, Rhee SG, Kim K and Chae HZ (2008): Irreversible oxidation of the active-site cysteine of peroxiredoxin to cysteine sulfonic acid for enhanced molecularchaperone activity. J. Biol. Chem 283:28873-28880.

Lindskog S (1960): Purification and properties of bovine erythrocyte carbonic anhydrase. Biochim Biophys Acta 39:218-226.

Link AJ, Robison K and Church GM (1997): Comparing the predicted and observed properties of proteins encoded in the genome of Escherichia coli K-12. Electrophoresis 18:1259–1313.

Linskey ME and Gilbert MR (1995): Glial differentiation: a review with implications for new directions in neuro-oncology. Neurosurgery 36:1-21.

Loncaster JA, Harris AL, Davidson SE, Logue JP, Hunter RD, Wycoff CC, Pastorek J, Ratcliffe PJ, Stratford IJ and West CM (2001): Carbonic anhydrase (CA IX) expression, a potential new intrinsic marker of hypoxia: correlations with tumor oxygen measurements and prognosis in locally advanced carcinoma of the cervix. Cancer Res 61:6394-6399.

Long H, Crean CD, Lee WH, Cummings OW and Gabig TG (2001): Expression of Clostridium perfringens enterotoxin receptors claudin-3 and claudin-4 in prostate cancer epithelium. Cancer Res 61:7878-7881.

92

Louis DN, Ohgaki H, Wiestler O, and Cavenee W (eds) (2007): WHO classification of tumors of the central nervous system. Lyon: International Agency for Research on Cancer, Press.

Lu KH, Patterson AP, Wang L, Marquez RT, Atkinson EN, Baggerly KA, Ramoth LR, Rosen DG, Liu J, Hellstrom I, Smith D, Hartmann L, Fishman D, Berchuck A, Schmandt R, Whitaker R, Gershenson DM, Mills GB and Bast RC Jr (2004): Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent patition analysis. Clin Cancer Res 10:3291-3300.

Maris JM, Courtright J, Houghton PJ, Morton CL, Gorlick R, Kolb EA, Lock R, Tajbakhsh M, Reynolds CP, Keir ST, Wu J and Smith MA (2008): Initial testing of the VEGFR inhibitor AZD2171 by the pediatric preclinical testing program. Pediatr Blood Cancer 50581-50587.

Martin TA and Jiang WG (2001): Tight junctions and their role in cancer metastasis. Histol Histopathol 16:1183-1195.

Martinez-Estrada OM, Culleres A, Soriano FX, Peinado H, Bolós V, Martínez FO, Reina M, Cano A, Fabre M and Vilaró S (2006): The transcription factors Slug and Snail act as repressors of Claudin-1 expression in epithelial cells. Biochem J 394:449-457.

Matsui M, Oshima M, Oshima H, Takaku K, Maruyama T, Yodoi J and Taketo MM (1996): Early embryonic lethality caused by targeted disruption of the mouse thioredoxin gene. Dev Biol 178:179-185.

Mazzon E and Cuzzocrea S (2007): Role of TNF-alpha in lung tight junction alteration in mouse model of acute lung inflammation. Respir Res 8:75.

McBride SM, Daganzo SM, Banerjee A, Gupta N, Lamborn KR, Prados MD, Berger MS, Wara WM and Haas-Kogan DA (2008): Radiation is an important component of multimodality therapy for pediatric non-pineal supratentorial primitive neuroectodermal tumors. Int J Radiat Oncol Biol Phys 72:1319-1323.

McCord JM and Fridovich I (1969): Superoxide dismutase. An enzymatic function for erythrocuprein (hemocuprein). J. Biol. Chem 244:6049-6055. 93

McKiernan JM, Buttyan R, Bander NH, Stifelman MD, Katz AE, Chen MW, Olsson CA and Sawczuk IS (1997): Expression of the tumor-associated gene MN: a potential biomarker for human renal cell carcinoma. Cancer Res 57:2362-2365.

Meldrum NU and Roughton FJ (1933): Carbonic anhydrase. Its preparation and properties. J Physiol 80:113-142.

Merikallio H, Kaarteenaho R, Pääkkö P, Lehtonen S, Hirvikoski P, Mäkitaro R, Harju T and Soini Y (2011): Zeb1 and twist are more commonly expressed in metastatic than primary lung tumours and show inverse associations with claudins. J Clin Pathol 64:136-140.

Merkle FT, Tramontin AD, Garcia-Verdugo JM and Alvarez-Buylla A (2004): Radial glia give rise to adult neural stem cells in the subventricular zone Proc Natl Acad Sci USA 101:17528-17532.

Mettler FA and Upton AC (2008): Medical Effects of Ionizing Radiation. Philadelphia, PA: Saunders Elsvier.

Michl P, Buchholz M, Rolke M, Kunsch S, Löhr M, McClane B, Tsukita S, Leder G, Adler G and Gress TM (2001): Claudin-4: a new target for pancreatic cancer treatment using Clostridium perfringens enterotoxin. Gastroenterology 121:678-684.

Mikheeva SA, Mikheev AM, Petit A, Beyer R, Oxford RG, Khorasani L, Maxwell JP, Glackin CA, Wakimoto H, González-Herrero I, Sánchez-García I, Silber JR, Horner PJ and Rostomily RC (2010): TWIST1 promotes invasion through mesenchymal change in human glioblastoma. Mol Cancer 9:194.

Mineta K, Yamamoto Y, Yamazaki Y, Tanaka H, Tada Y, Saito K, Tamura A, Igarashi M, Endo T, Takeuchi K and Tsukita S (2011): Predicted expansion of the claudin multigene family. FEBS Lett 585:606-612.

Mitic LL and Anderson JM (1998): Molecular architecture of tight junctions. Annu Rev Physiol 60:121-142.

94

Modena P, Lualdi E, Facchinetti F, Veltman J, Reid JF, Minardi S, Janssen I, Giangaspero F, Forni M, Finocchiaro G, Genitori L, Giordano F, Riccardi R, Schoenmakers EF, Massimino M and Sozzi G (2006): Identification of tumor-spesific molecular signatures in intracranial ependymoma and association with clinical charasteristics. J Clin Oncol 24:5223-5233.

Monari M, Foschi J, Calabrese C, Liguori G, Di Febo G, Rizzello F, Gionchetti P, Trinchero A and Serrazanetti GP (2009): Implications of antioxidant enzymes in human gastric neoplasms. Int J Mol Med 24:693-700.

Morin PJ (2005): Claudin proteins in human cancer: promising new targets for diagnosis and therapy. Cancer Res 65:9603-9606.

Morita KH, Sasaki K, Fujimoto M, Furuse S and Tsukita (1999a): Claudin-11/OSP-based tight junctions in myelinated sheaths of oligodendrocytes and Sertoli cells in testis. J Cell Biol 145:579588.

Morita KH, Sasaki M, Furuse S and Tsukita (1999b): Endothelial claudin: claudin-5/TMVCF constitutes tight junction strands in endothelial cellsJ. Cell Biol 147:185-194

Morita K, Sasaki H, Furuse K, Furuse M, Tsukita S and Miyachi Y (2003): Expression of claudin-5 in dermal vascular endothelia. Exp Dermatol 12:289-295.

Mueller S and Chang S (2009): Pediatric Brain tumors: Current Treatment Strategies and Future Therapeutic Approaches Neurotherapeutics 6:570-586.

Mullin JM (1997): Potential interplay between luminal growth factors and increased tight junction permeability in epithelial carcinogenesis. J Exp Zool 279:484-489.

Nagy Z, Peters H and Hüttner I (1984): Fracture faces of cell junctions in cerebral endothelium during normal and hyperosmotic conditions. Lab Invest 50:313-322.

Nakai H, Byers MG, Venta PJ, Tashian RE and Shows TB (1987): The gene for human carbonic anhydrase II (CA2) is located at chromosome 8q22. Cytogenet Cell Genet 44:234-235.

95

Nakamura H, Herzenberg LA, Bai J, Araya S, Kondo N, Nishinaka Y, Herzenberg LA and Yodoi J (2001): Circulating thioredoxin suppresses lipopolysaccharide-induced neutrophil chemotaxis. Proc Natl Acad Sci U S A 98:15143-15148.

Neumann CA, Krause DS, Carman CV, Das S, Dubey DP, Abraham JL, Bronson RT, Fujiwara Y, Orkin SH and Van Etten RA (2003): Essential role for the peroxiredoxin Prdx1 in erythrocyte antioxidant defence and tumour suppression. Nature 424:561-565.

Nichols LS, Ashfaq R and Iacobuzio-Donahue CA (2004): Claudin 4 protein expression in primary and metastatic pancreatic cancer: support for use as a therapeutic target. Am J Clin Pathol 121:226230.

Niimi T, Nagashima K, Ward JM, Minoo P, Zimonjic DB, Popescu NC and Kimura S (2001): claudin-18, a novel downstream target gene for the T/EBP/NKX2.1 homeodomain transcription factor, encodes lung- and stomach-specific isoforms through alternative splicing. Mol Cell Biol 21:7380-7390.

Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, Furuse M and Tsukita S (2003): Sizeselective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol 161:653-660.

Noh DY, Ahn SJ, Lee RA, Kim SW, Park IA and Chae HZ (2001): Overexpression of peroxiredoxin in human breast cancer. Anticancer Res 21:2085-2090.

Noor R, Mittal S and Iqbal J (2002): Superoxide dismutase--applications and relevance to human diseases. Med Sci Monit. 2002;8:RA210-5.

Northcott PA, Fernandez LA, Hagan JP, Ellison DW, Grajkowska W, Gillespie Y, Grundy R, Van Meter T, Rutka JT, Croce CM, Kenney AM and Taylor MD (2009): The miR-17/92 polycistron is up-regulated in sonic hedgehog-driven medulloblastomas and induced by N-myc in sonic hedgehog-treated cerebellar neural precursors. Cancer Res 69:3249–3255.

Northcott PA, Korshunov A, Witt H, Hielscher T, Eberhart CG, Mack S, Bouffet E, Clifford SC, Hawkins CE, French P, Rutka JT, Pfister S and Taylor MD (2011): Medulloblastoma comprises four distinct molecular variants. J Clin Oncol 29:1408-1414. 96

Novoselov SV, Peshenko IV, Popov VI, Novoselov VI, Bystrova MF, Evdokimov VJ, Kamzalov SS, Merkulova MI, Shuvaeva TM, Lipkin VM and Fesenko EE (1999): Localization of 28-kDa peroxiredoxin in rat epithelial tissues and its antioxidant properties. Cell Tissue Res. 1999;298:471480.

Ohgaki H and Kleihues P (2005): Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol 64:479-489.

Okado-Matsumoto A, Matsumoto A, Fujii J and Taniguchi N (2000): Peroxiredoxin IV is a secretable protein with heparin-binding properties under reduced conditions. J Biochem 127:493501.

Oliveira SS and Morgado-Diaz JA (2007): Claudins: Multifunctional Players in Epithelial Tight Junctions and Their Role in Cancer. Cell Mol Life Sci 64:17-28.

Opavský R, Pastoreková S, Zelník V, Gibadulinová A, Stanbridge EJ, Závada J, Kettmann R and Pastorek J (1996): Human MN/CA9 gene, a novel member of the carbonic anhydrase family: structure and exon to protein domain relationships. Genomics 33:480-487.

Packer RJ (1999): Childhood medulloblastoma: progress and future challenges. Brain Dev 21:7581.

Palma L, Russo A and Mercuri S (1983): Cystic cerebral astrocytomas in infancy and childhood: long-term results. Childs Brain 10:79-91.

Park SH, Chung YM, Lee YS, Kim HJ, Kim JS, Chae HZ and Yoo YD (2000): Antisense of human peroxiredoxin II enhances radiation-induced cell death. Clin Cancer Res 6:4915-4920.

Parkkila AK, Parkkila S, Juvonen T and Rajaniemi H (1993): Carbonic anhydrase isoenzymes II and I are present in the zona glomerulosa cells of the human adrenal gland. Histochemistry 99:3741.

Parkkila AK, Herva R, Parkkila S and Rajaniemi H (1995): Immunohistochemical demonstration of human carbonic anhydrase isoenzyme II in brain tumours. Histochem J 27:974-982. 97

Parkkila S and Parkkila AK (1996): Carbonic anhydrase in the alimentary tract. Roles of the different isozymes and salivary factors in the maintenance of optimal conditions in the gastrointestinal canal. Scand J Gastroenterol 31:305-317.

Pastorek J, Pastoreková S, Callebaut I, Mornon JP, Zelník V, Opavský R, Zat'ovicová M, Liao S, Portetelle D, Stanbridge EJ and et al. (1994): Cloning and characterization of MN, a human tumorassociated protein with a domain homologous to carbonic anhydrase and a putative helix-loop-helix DNA binding segment. Oncogene 9:2877-2888.

Pastoreková S, Závadová Z, Kostál M, Babusíková O and Závada J (1992): A novel quasi-viral agent, MaTu, is a two-component system. Virology 187:620-626.

Pastoreková S, Parkkila S, Parkkila AK, Opavský R, Zelník V, Saarnio J and Pastorek J (1997): Carbonic anhydrase IX, MN/CA IX: analysis of stomach complementary DNA sequence and expression in human and rat alimentary tracts. Gastroenterology 112:398-408.

Pastoreková S, Parkkila S, Pastorek J and Supuran CT (2004): Carbonic anhydrases: current state of the art, therapeutic applications and future prospects. J Enzyme Inhib Med Chem 19:199-229.

Patard JJ, Fergelot P, Karakiewicz PI, Klatte T, Trinh QD, Rioux-Leclercq N, Said JW, Belldegrun AS and Pantuck AJ (2008): Low CAIX expression and absence of VHL gene mutation are associated with tumor aggressiveness and poor survival of clear cell renal cell carcinoma. Int J Cancer 123:395-400.

Peshenko IV, Novoselov VI, Evdokimov VA, Nikolaev YuV, Shuvaeva TM, Lipkin VM and Fesenko EE (1996): Novel 28-kDa secretory protein from rat olfactory epithelium. FEBS Lett 381:12-14.

Peshenko IV, Novoselov VI, Evdokimov VA, Nikolaev YV, Kamzalov SS, Shuvaeva TM, Lipkin VM and Fesenko EE (1998): Identification of a 28 kDa secretory protein from rat olfactory epithelium as a thiol-specific antioxidant. Free Radic Biol Med 25:654-659.

98

Pfister S, Janzarik WG, Remke M, Ernst A, Werft W, Becker N, Toedt G, Wittmann A, Kratz C, Olbrich H, Ahmadi R, Thieme B, Joos S, Radlwimmer B, Kulozik A, Pietsch T, Herold-Mende C, Gnekow A, Reifenberger G, Korshunov A, Scheurlen W, Omran H and Lichter P (2008): BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas. J Clin Invest 118:1739-1749.

Plaisant F, Clippe A, Vander Stricht D, Knoops B and Gressens P (2003): Recombinant peroxiredoxin 5 protects against excitotoxic brain lesions in newborn mice. Free Radic Biol Med 34:862-872.

Pokhrel A and Hakulinen T (2009): Age-standardisation of relative survival ratios of cancer patients in a comparison between countries, genders and time periods. Eur J Cancer 45:642-647.

Power JH, Asad S, Chataway TK, Chegini F, Manavis J, Temlett JA, Jensen PH, Blumbergs PC and Gai WP (2008): Peroxiredoxin 6 in human brain: molecular forms, cellular distribution and association with Alzheimer's disease pathology. Acta Neuropathol 115:611-622.

Powis G and Montfort WR (2001): Properties and biological activities of thioredoxins. Annu Rev Pharmacol Toxicol 41:261-295.

Prayson RA (1999): Clinicopathologic study of 61 patients with ependymoma including MIB-1 immunohistochemistry. Ann Diagn Pathol 3:11-18.

Proescholdt MA, Mayer C, Kubitza M, Schubert T, Liao SY, Stanbridge EJ, Ivanov S, Oldfield EH, Brawanski A and Merrill MJ (2005): Expression of hypoxia-inducible carbonic anhydrases in brain tumors. Neuro Oncol 7:465–475.

Rabilloud T, Heller M, Gasnier F, Luche S, Rey C, Aebersold R, Benahmed M, Louisot P and Lunardi J (2002): Proteomics analysis of cellular response to oxidative stress. Evidence for in vivo overoxidation of peroxiredoxins at their active site. J Biol Chem 277:19396-19401.

Raffel J, Bhattacharyya AK, Gallegos A, Cui H, Einspahr JG, Alberts DS and Powis G (2003): Increased expression of thioredoxin-1 in human colorectal cancer is associated with decreased patient survival. J Lab Clin Med 142:46-51. 99

Rahner C, Mitic LL and Anderson JM (2001): Heterogeneity in expression and subcellular localization of claudins 2, 3, 4, and 5 in the rat liver, pancreas, and gut. Gastroenterol 120:411-422.

Rand V, Prebble E, Ridley L, Howard M, Wei W, Brundler MA, Fee BE, Riggins GJ, Coyle B and Grundy RG; Children's Cancer and Leukaemia Group Biological Studies Committee (2008): Investigation of chromosome 1q reveals differential expression of members of the S100 family in clinical subgroups of intracranial paediatric ependymoma. Br J Cancer 99:1136-1143.

Rappa G, Gamcsik MP, Mitina RL, Baum C, Fodstad O and Lorico A (2003): Retroviral transfer of MRP1 and gamma-glutamyl cysteine synthetase modulates cell sensitivity to L-buthionine-S,Rsulphoximine (BSO): new rationale for the use of BSO in cancer therapy. Eur J Cancer 39:120–128.

Reese T and Karnovsky M (1967): Fine structural localization of a blood-brain barrier to exogenous proxidase. J Cell Biol 34:207-217.

Reimers TS, Ehrenfels S, Mortensen EL, Schmiegelow M, Sønderkaer S, Carstensen H, Schmiegelow K and Müller J (2003): Cognitive deficits in long-term survivors of childhood brain tumors: Identification of predictive factors. Med Pediatr Oncol 40:26-34.

Reni M, Gatta G, Mazza E and Vecht C (2007): Ependymoma. Crit Rev Oncol Hematol 63:81-89.

Reyes JL, Lamas M, Martin D, del Carmen Namorado M, Islas S, Luna J, Tauc M and GonzálezMariscal L (2002): The renal segmental distribution of claudins changes with development. Kidney Int 62:476-487.

Risau W and Wolburg H (1990): Development of the blood-brain barrier. Trends Neurosci 13:174178.

Roberts RO, Lynch CF, Jones MP and Hart MN (1991): Medulloblastoma: a polpulation-based study of 532 cases. J Neuropathol Exp Neurol 50:134-144.

Robertson N, Potter C and Harris AL (2004): Role of carbonic anhydrase IX in human tumor cell growth, survival, and invasion. Cancer Res 64:6160-6165.

100

Robertson PL, Zeltzer PM, Boyett JM, Rorke LB, Allen JC, Geyer JR, Stanley P, Li H, Albright AL, McGuire-Cullen P, Finlay JL, Stevens KR Jr, Milstein JM, Packer RJ and Wisoff J (1998): Survival and prognostic factors following radiation therapy and chemotherapy for ependymomas in children: a report of the Children's Cancer Group. J Neurosurg 88:695-703.

Saarnio J, Parkkila S, Parkkila AK, Waheed A, Casey MC, Zhou XY, Pastoreková S, Pastorek J, Karttunen T, Haukipuro K, Kairaluoma MI and Sly WS (1998): Immunohistochemistry of carbonic anhydrase isozyme IX (MN/CA IX) in human gut reveals polarized expression in the epithelial cells with the highest proliferative capacity. J Histochem Cytochem 46:497-504.

Saarnio J, Parkkila S, Parkkila AK, Pastoreková S, Haukipuro K, Pastorek J, Juvonen T and Karttunen TJ (2001): Transmembrane carbonic anhydrase, MN/CA IX, is a potential biomarker for biliary tumours. J Hepatol 35:643-649.

Sadek CM, Jiménez A, Damdimopoulos AE, Kieselbach T, Nord M, Gustafsson JA, Spyrou G, Davis EC, Oko R, van der Hoorn FA and Miranda-Vizuete A (2003): Characterization of human thioredoxin-like 2. A novel microtubule-binding thioredoxin expressed predominantly in the cilia of lung airway epithelium and spermatid manchette and axoneme. J Biol Chem 278:13133-13142.

Sallinen PK, Haapasalo HK, Visakorpi T, Helén PT, Rantala IS, Isola JJ and Helin HJ (1994): Prognostication of astrocytoma patient survival by Ki-67 (MIB-1), PCNA, and S-phase fraction using archival paraffin-embedded samples. J Pathol 174:275-282.

Sandström J, Nilsson P, Karlsson K and Marklund SL (1994): 10-fold increase in human plasma extracellular superoxide dismutase content caused by a mutation in heparin-binding domain. J Biol Chem 269:19163-19166.

Sarafian TA, Verity MA, Vinters HV, Shih CC, Shi L, Ji XD, Dong L and Shau H (1999): Differential expression of peroxiredoxin subtypes in human brain cell types. J Neurosci Res 56:206212.

Sasagawa I, Matsuki S, Suzuki Y, Iuchi Y, Tohya K, Kimura M, Nakada T and Fujii J (2001): Possible involvement of the membrane-bound form of peroxiredoxin 4 in acrosome formation during spermiogenesis of rats. Eur J Biochem 268:3053-3061. 101

Schiffer D and Giordana MT (1998): Prognosis of ependymoma. Childs Nerv Syst 14:357-361.

Schiffer D, Chiò A, Giordana MT, Migheli A, Palma L, Pollo B, Soffietti R and Tribolo A (1991): Histologic prognostic factors in ependymoma. Childs Nerv Syst 7:177-182.

Seo MS, Kang SW, Kim K, Baines IC, Lee TH and Rhee SG (2000): Identification of a new type of mammalian peroxiredoxin that forms an intramolecular disulfide as a reaction intermediate. J Biol Chem 275:20346-20354.

Shau H, Gupta RK and Golub SH (1993): Identification of a natural killer enhancing factor (NKEF) from human erythroid cells. Cell. Immunol 147:1-11.

Shau H, Butterfield LH, Chiu R and Kim A (1994): Cloning and sequence analysis of candidate human natural killer-enhancing factor genes. Immunogenetics 40:129-134.

Shepherd AG, Manson MM, Ball HW and McLellan LI (2000): Regulation of rat glutamatecysteine ligase (gamma-glutamylcysteine synthetase) subunits by chemopreventive agents and in aflatoxin B(1)-induced preneoplasia. Carcinogenesis 21:1827-1834.

Sierra-Rivera E, Summar ML, Dasouki M, Krishnamani MR, Phillips JA and Freeman ML (1995): Assignment of the gene (GLCLC) that encodes the heavy subunit of gamma-glutamylcysteine synthetase to human chromosome 6. Cytogenet Cell Genet 70:278-279. Sierra-Rivera E, Dasouki M, Summar ML, Krishnamani MR, Meredith M, Rao PN, Phillips JA 3 rd and Freeman ML (1996): Assignment of the human gene (GLCLR) that encodes the regulatory subunit of gamma-glutamylcysteine synthetase to chromosome 1p21. Cytogenet Cell Genet 72:252254.

Singh AB, Sharma A and Dhawan P (2010): Claudin family of proteins and cancer: an overview. J Oncol 541957.

Sly WS and Hu PY (1995): Human carbonic anhydrases and carbonic anhydrase deficiencies. Annu Rev Biochem 64:375-401.

102

Sly WS, Hewett-Emmett D, Whyte MP, Yu YS and Tashian RE (1983): Carbonic anhydrase II deficiency identified as the primary defect in the autosomal recessive syndrome of osteopetrosis with renal tubular acidosis and cerebral calcification. Proc Natl Acad Sci U S A 80:2752-2756.

Soini Y (2005): Expression of claudins 1, 2, 3, 4, 5 and 7 in various types of tumours. Histopathology 46:551-560.

Soini Y, Vakkala M, Kahlos K, Pääkkö P and Kinnula V (2001): MnSOD expression is less frequent in tumour cells of invasive breast carcinomas than in in situ carcinomas or non-neoplastic breast epithelial cells. J Pathol 195:156-162.

Soini Y, Karihtala P, Mäntyniemi A, Turunen N, Pääkkö P and Kinnula V (2004): Glutamate-Lcysteine ligase in breast carcinomas. Histopathology 44:129-135.

Soini Y, Kinnula V, Kahlos K and Pääkkö P (2006a): Claudins in differential diagnosis between mesothelioma and metastatic adenocarcinoma of the pleura. J Clin Pathol. 2006;59:250-4

Soini Y, Tommola S, Helin H and Martikainen P (2006b): Claudins 1, 3, 4 and 5 in gastric carcinoma, loss of claudin expression associates with the diffuse subtype. Virchows Arch 448:5258.

Soler AP, Miller RD, Laughlin KV, Carp NZ, Klurfeld DM and Mullin JM (1999): Increased tight junctional permeability is associated with the development of colon cancer. Carcinogenesis 20:1425-1431.

Soma T, Chiba H, Kato-Mori Y, Wada T, Yamashita T, Kojima T and Sawada N (2004): Thr(207) of claudin-5 is involved in size-selective loosening of the endothelial barrier by cyclic AMP. Exp Cell Res 300:202-212.

Statistics Finland (2011). Statistical databases for cause of death in years 2003-2009.

St Clair DK, Wan XS, Oberley TD, Muse KE and St Clair WH (1992): Suppression of radiationinduced neoplastic transformation by overexpression of mitochondrial superoxide dismutase. Mol Carcinog 6:238-242. 103

Streicher KL, Sylte MJ, Johnson SE and Sordillo LM (2004): Thioredoxin reductase regulates angiogenesis by increasing endothelial cell-derived vascular entohelial growth factor. Nutr Cancer 50:221-231.

Sumida Y, Nakashima T, Yoh T, Nakajima Y, Ishikawa H, Mitsuyoshi H, Sakamoto Y, Okanoue T, Kashima K, Nakamura H and Yodoi J (2000): Serum thioredoxin levels as an indicator of oxidative stress in patients with hepatitis C virus infection. J Hepatol 33:616-622.

Sun QA, Kirnarsky L, Sherman S and Gladyshev VN (2001): Selenoprotein oxidoreductase with specificity for thioredoxin and glutathione systems. Proc Natl Acad Sci U S A 98:3673-3678.

Supuran CT (2004): Carbonic anhydrases: Catalytic mechanism, distribution and physiological roles. In: Carbonic Anhydrase: Its Inhibitors ans Activators. CT Supuran, A Scozzafava, J Conway (eds), pp. 1-24, CRC Press Boca Raton-London-New York-Washington D.C.

Supuran CT, Casini A and Scozzafava A (2003): Protease inhibitors of the sulfonamide type: anticancer, antiinflammatory, and antiviral agents. Med Res Rev 23:535-558.

Svastová E, Zilka N, Zat'ovicová M, Gibadulinová A, Ciampor F, Pastorek J and Pastoreková S (2003): Carbonic anhydrase IX reduces E-cadherin-mediated adhesion of MDCK cells via interaction with beta-catenin. Exp Cell Res 290:332-345.

Svensk AM, Soini Y, Pääkkö P, Hiravikoski P and Kinnula VL (2004): Differential expression of superoxide dismutases in lung cancer. Am J Clin Pathol 122:395-404.

Takala H, Saarnio J, Wiik H and Soini Y (2007): Claudins 1, 3, 4, 5 and 7 in esophageal cancer: loss of claudin 3 and 4 expression is associated with metastatic behavior. APMIS 115:838-847.

Tamimi RM, Hankinson SE, Spiegelman D, Colditz GA and Hunter DJ (2004): Manganese superoxide dismutase polymorphism, plasma antioxidants, cigarette smoking, and risk of breast cancer. Cancer Epidemiol Biomarkers Prev 13:989–996.

Tao-Cheng JH, Nagy Z and Brightman MW (1987): Tight junctions of brain endothelium in vitro are enhanced by astroglia. J Neurosci 7:3293-3299. 104

Tashian RE (1992): Genetics of the mammalian carbonic anhydrases. Adv Genet 30:321-356.

Tatum JL, Kelloff GJ, Gillies RJ, Arbeit JM, Brown JM, Chao KS, Chapman JD, Eckelman WC, Fyles AW, Giaccia AJ, Hill RP, Koch CJ, Krishna MC, Krohn KA, Lewis JS, Mason RP, Melillo G, Padhani AR, Powis G, Rajendran JG, Reba R, Robinson SP, Semenza GL, Swartz HM, Vaupel P, Yang D, Croft B, Hoffman J, Liu G, Stone H and Sullivan D (2006): Hypoxia: importance in tumor biology, noninvasive measurement by imaging, and value of its measurement in the management of cancer therapy. Int J Radiat Biol 82:699-757.

Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, Hartwell K, Onder TT, Gupta PB, Evans KW, Hollier BG, Ram PT, Lander ES, Rosen JM, Weinberg RA and Mani SA (2010): Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc Natl Acad Sci U S A 107:15449-15454.

Taylor MD, Poppleton H, Fuller C, Su X, Liu Y, Jensen P, Magdaleno S, Dalton J, Calabrese C, Board J, Macdonald T, Rutka J, Guha A, Gajjar A, Curran T, Gilbertson RJ (2005): Radial glia cells are candidate stem cells of ependymoma. Cancer Cell 8:323-335.

Teicher BA (1994): Hypoxia and drug resistance. Cancer Metastasis Rev 13:139-168.

Tew KD (1994): Glutathione-associated enzymes in anticancer drug resistance. Cancer Res 54:4313-4320.

Thiery JP, Acloque H, Huang RY and Nieto MA (2009): Epithelial-mesenchymal transitions in development and disease. Cell 139:871-890.

Tipnis SR, Blake DG, Shepherd AG and McLellan LI (1999): Overexpression of the regulatory subunit of c-glutamylcysteine synthetase in HeLa cells increases c- glutamylcysteine synthetase activity and confers drug resistance. Biochem. J 337:559–566.

Tomlinson FH, Scheithauer BW, Hayostek CJ, Parisi JE, Meyer FB, Shaw EG, Weiland TL, Katzmann JA and Jack CR Jr (1994): The significance of atypia and histologic malignancy in pilocytic astrocytoma of the cerebellum: a clinicopathologic and flow cytometric study. J Child Neurol 9:301-310. 105

Trush MA and Kensler TW (1991): An overview of the relationship between oxidative stress and chemical carcinogenesis. Free Radic Biol Med 10:201-209.

Tsukita S and Furuse M (2000): Poresis in the wall: claudins constitute tight junction strands containing aqueous pores. J CEll Biol 149:13-16.

Tsukita S, Furuse M and Itoh M (2001): Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol 2:285-293.

Turner JR, Odze RD, Crum CP and Resnick MB (1997): MN antigen expression in normal, preneoplastic, and neoplastic esophagus: a clinicopathological study of a new cancer-associated biomarker. Hum Pathol 28:740-744.

Türeci O, Sahin U, Vollmar E, Siemer S, Göttert E, Seitz G, Parkkila AK, Shah GN, Grubb JH, Pfreundschuh M and Sly WS (1998): Human carbonic anhydrase XII: cDNA cloning, expression, and chromosomal localization of a carbonic anhydrase gene that is overexpressed in some renal cell cancers. Proc Natl Acad Sci U S A 95:7608-7613.

Turunen M, Talvensaari-Mattila A, Soini Y and Santala M (2009): Claudin-5 overexpression correlates with aggressive behavior in serous ovarian adenocarcinoma. Anticancer Res 29:51855189.

Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M and Telser J (2007): Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol 39:44–84. van Meer G, Gumbiner B and Simons K (1986): The tight junction does not allow lipid molecules to diffuse from one epithelial cell to the next. Nature 322:639-641.

Vinchon M, Leblond P, Noudel R and Dhellemmes P (2005): Intracranial ependymomas in childhood: recurrence, reoperation, and outcome. Childs Nerv Syst 21:221-226.

Venkatramani R, Dhall G, Patel M, Grimm J, Hawkins C, McComb G, Krieger M, Wong K, O'Neil S, Finlay JL (2011): Supratentorial ependymoma in children: To observe or to treat following gross total resection? Pediatr Blood Cancer 2011 Mar 2. [Epub ahead of print]. 106

Vermylen P, Roufosse C, Burny A, Verhest A, Bosschaerts T, Pastorekova S, Ninane V and Sculier JP (1999): Carbonic anhydrase IX antigen differentiates between preneoplastic malignant lesions in non-small cell lung carcinoma. Eur Respir J 14:806-811.

Waldron JS and Tihan T (2003): Epidemiology and pathology of intraventricular tumors. Neurosurg Clin N Am 14:469-482.

Wang HQ, Du ZX, Liu BQ, Gao YY, Meng X, Guan Y and Zhang HY (2009): TNF-related apoptosis-inducing ligand suppresses PRDX4 expression. FEBS Lett 583:1511-1515.

Watson GA, Kadota RP and Wisoff JH (2001): Multidisciplinary management of pediatric lowgrade gliomas. Semin Radiat Oncol 11:152-162.

Weinstein RS, Merk FB and Alroy J (1976): The structure and function of intercellular junctions in cancer. Adv Cancer Res 23:23-89.

Welsh SJ, Bellamy WT, Briehl MM and Powis G (2002): The redox protein thioredoxin-1 (Trx-1) increases hypoxia-inducible factor 1alpha protein expression: Trx-1 overexpression results in increased vascular endothelial growth factor production and enhanced tumor angiogenesis. Cancer Res 62:5089-5095.

Witz G (1991): Active oxygen species as factors in multistage carcinogenesis. Proc Soc Exp Biol Med 198:675-682.

Wonsey DR, Zeller KI and Dang CV (2002): The c-Myc target gene PRDX3 is required formitochondrial homeostasis and neoplastic transformation. Proc Natl Acad Sci U S A 99:66496654.

Wood ZA, Schroder E, Robin Harris J and Poole LB (2003): Structure, mechanism and regulation of peroxiredoxins.Trends Biochem. Sci 28:32–40.

Wu XY, Fu ZX and Wang XH (2010): Peroxiredoxins in colorectal neoplasms. Histol Histopathol 25:1297-1303.

107

Wykoff CC, Beasley NJ, Watson PH, Turner KJ, Pastorek J, Sibtain A, Wilson GD, Turley H, Talks KL, Maxwell PH, Pugh CW, Ratcliffe PJ and Harris AL (2000): Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res 60:7075-7083.

Wykoff CC, Beasley N, Watson PH, Campo L, Chia SK, English R, Pastorek J, Sly WS, Ratcliffe P and Harris AL (2001): Expression of the hypoxia-inducible and tumor-associated carbonic anhydrases in ductal carcinoma in situ of the breast. Am J Pathol 158:1011-1019.

Xu Y, Krisnan A, Wan XS, Majima H, Yeh CC, Ludewig G, Kasarskis EJ and St Clair DK (1999): Mutations in the promoter reveal a cause for the reduced expression of the human manganese superoxide dismutase gene in cancer cells. Oncogene18:93-102.

Yamada Y, Nakamura H, Adachi T, Sannohe S, Oyamada H, Kayaba H, Yodoi J and Chihara J (2003): Elevated serum levels of thioredoxin in patients with acute exacerbation of asthma. Immunol Lett 86:199-205.

Yamashita H, Avraham S, Jiang S, London R, Van Veldhoven PP, Subramani S, Rogers RA and Avraham H (1999): Characterization of human and murine PMP20 peroxisomal proteins that exhibit antioxidant activity in vitro. J. Biol. Chem 274:29897–29904.

Yan T, Oberley LW, Zhong W and St Clair DK (1996): Manganese-containing superoxide dismutase overexpression causes phenotypic reversion in SV40-transformed human lung fibroblasts. Cancer Res 56:2864-2871.

Yanagawa T, Ishikawa T, Ishii T, Tabuchi K, Iwasa S, Bannai S, Omura K, Suzuki H and Yoshida H (1999): Peroxiredoxin I expression in human thyroid tumors. Cancer Lett 145:127-132.

Yen G, Croci A, Dowling A, Zhang S, Zoeller RT and Darling DS (2001): Developmental and functional evidence of a role for Zfhep in neural cell development. Brain Res Mol Brain Res 96:5967.

Yin GY, Yin YF and He XF (1995): Effect of zhuchun pill on immunity and endocrine function of elderly with kidney-yang deficiency. Zhongguo Zhong Xi Yi Jie He Za Zhi 15:601-603. 108

Yoo MH, Xu XM, Carlson BA, Gladyshev VN and Hatfield DL (2006): Thioredoxin reductase 1 deficiency reverses tumor phenotype and tumorigenicity of lung carcinoma cells. J Biol Chem 281:13005-13008.

Yoshiura K, Nakaoka T, Nishishita T, Sato K, Yamamoto A, Shimada S, Saida T, Kawakami Y, Takahashi TA, Fukuda H, Imajoh-Ohmi S, Oyaizu N and Yamashita N (2005): Carbonic anhydrase II is a tumor vessel endothelium-associated antigen targeted by dendritic cell therapy. Clin Cancer Res 11:8201-8207.

Zacharoulis S and Moreno L (2009): Ependymoma: An Update. J. Child Neurol 24:1431-1438.

Zacharoulis S, Ji L, Pollack IF, Duffner P, Geyer R, Grill J, Schild S, Jaing TH, Massimino M, Finlay J and Sposto R (2008): Metastatic ependymoma: a multi-institutional retrospective analysis of prognostic factors. Pediatr Blood Cancer 50:231-235.

Závada J, Závadová Z, Pastorek J, Biesová Z, Jezek J and Velek J (2000): Human tumourassociated cell adhesion protein MN/CA IX: identification of M75 epitope and of the region mediating cell adhesion. Br J Cancer 82:1808-1813.

Zhang P, Liu B, Kang SW, Seo MS, Rhee SG and Obeid LM (1997): Thioredoxin peroxidase is a novel inhibitor of apoptosis with a mechanism distinct from that of Bcl-2. J Biol Chem 272:3061530618.

Zhang XZ, Xiao ZF, Li C, Xiao ZQ, Yang F, Li DJ, Li MY, Li F and Chen ZC (2009): Triosephosphate isomerase and peroxiredoxin 6, two novel serum markers for human lung squamous cell carcinoma. Cancer Sci 100:2396-2401.

Zhong W, Oberley LW, Oberley TD and St Clair DK (1997): Suppression of the malignant phenotype of human glioma cells by overexpression of manganese superoxide dismutase. Oncogene 14:481-490.

Zhu Y, Brännström M, Janson PO and Sundfeldt K (2006): Differences in expression patterns of the tight junction proteins,claudin 1, 3, 4 and 5, in human ovarian surface epithelium as compared to epithelia in inclusion cysts and epithelial ovarian tumours. Int J Cancer 118:1884-1891. 109

10

ORIGINAL COMMUNICATIONS I-IV

110

Clinical Neuropathology, Vol. 26 – No. n/2007 (nnn-nnn)

Peroxiredoxins and antioxidant enzymes in pilocytic astrocytomas Original ©2007 Dustri-Verlag Dr. K. Feistle ISSN 0722-5091

K. Nordfors1,2, J. Haapasalo1,2, P. Helén3, A. Paetau4, L. Paljärvi1, H. Kalimo4,5, V.L. Kinnula6, Y. Soini7 and H. Haapasalo1 1Department

of Pathology, Center for Laboratory Medicine, Tampere University Hospital, Tampere, 2Faculty of Medicine, University of Turku, 3Unit of Neurosurgery, Tampere University Hospital, Tampere, 4Department of Pathology, Helsinki University Hospital, Helsinki, Finland, 5Department of Neuropathology, University of Uppsala, Sweden, 6Department of Medicine, Division of Pulmonary Medicine, University of Helsinki, 7Department of Pathology, Oulu University Hospital, Oulu, Finland Peroxiredoxins in pilocytic astrocytomas

Key words antioxidant – antioxidative enzymes – peroxiredoxins – pilocytic astrocytoma – prognosis

Received August 13, 2006; accepted in revised form January 9, 2007 Correspondence to K. Nordfors, MB Department of Pathology, Center for Laboratory Medicine, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland [email protected].

Abstract. Objective: Peroxiredoxins are antioxidant enzymes (AOEs), which are redox-regulated thiol proteins with potential effects on the growth, invasion and drug resistance of neoplastic cells. In this study, their biology and clinical significance were examined in pilocytic astrocytomas (PAs). Material and methods: The expression of peroxiredoxins (Prx I-VI) was investigated in 105 PAs by the means of immunohistochemistry and compared with the expression of selected other antioxidant enzymes, cell proliferation, angiogenesis, apoptosis, p53, histopathology and patient survival. Results: Peroxiredoxins were strongly expressed in general suggesting that oxidative damage and consequent defense takes place during the progression of pilocytic astrocytomas. In agreement with this hypothesis, several other AOEs correlated with the degenerative features and angiogenesis possibly associated with reactive oxygen species-derived cellular damage. Moreover, the expression of the AOEs was associated with each other indicating a concurrent activation of the enzymes. With the exception of manganese superoxide dismutase (MnSOD), a strong expression of AOEs was generally associated with higher cell proliferation. Prx VI seemed to have a positive association with a longer recurrence-free interval while other AOEs had no association with patient survival. Many AOEs, such as MnSOD, induce chemo- and radioresistance and are highly elevated in aggressive malignancies. PAs lack this confounding factor, and these tumors are treated only by surgery. Conclusions: Taken together, the results of this study on pilocytic astrocytomas suggest that the levels of Prxs and other AOEs and their related thiol proteins are generally strongly expressed in these tumors. At least Prx VI can contribute to tumor behavior which can make it a potential prognostic factor.

Introduction Antioxidant enzymes (AOEs) regulate the cellular redox state and constitute the major cellular protection factors against reactive oxygen species (ROS). These (e.g. O2–, H2O2, OH–) are harmful, since they cause oxidative damage to cellular proteins, lipids and genetic material. Low levels of ROS also modulate cell proliferation and apoptosis, and activate/induce the synthesis of growth factors [Kinnula et al. 2004]. Superoxide dismutases (SODs), glutathione-related enzyme systems such as glutamate cysteine ligase and catalase are the major antioxidant enzymes in mammalian cells. In addition to these, other major regulators of the cellular redox state include the thioredoxin (Trx) and peroxiredoxin (Prx) systems. These enzymes are also found in malignant tumors, inducing resistance of tumor cells to cytotoxic drugs and radiation [Kinnula and Crapo 2004], but possibly also independently contributing to tumor growth and invasion. Prxs occur in a wide variety of organisms from prokaryotes to mammals. They are present in various cellular compartments and reduce peroxides to the corresponding alcohol (or water), just like the other peroxidases. However, the Prx family differs in some respects from other groups of peroxidases and other antioxidant enzymes. Peroxiredoxins act both as a cosubstrates and peroxidases. Each Prx has a unique function and also interacts with other family members. They are expressed in different cell compartments in-

NP-6036 / 22.03.2007

2

Nordfors, Haapasalo, Helén et al.

cluding the cytosol, nucleus and mitochondria and also extracellularly [Fujii and Ikeda 2002]. Prx I and II are cytosolic proteins, Prx III is present in mitochondria and Prx IV in lysosomes, endoplasmic reticulum and extracellularly. Prx V is located in peroxisomes and mitochondria, and because of its location it is thought to have a more important role in protection against ROS than other Prxs. Prx VI is found at high concentrations, especially in the lung [Kinnula et al. 2002a]. Prxs are also expressed in the brain. In normal brain tissue, Prx I is expressed primarily in astrocytes and Prx II in neurons [Sarafian et al. 1999]. There are changes of Prx II expression, e.g. in Parkinson’s and Alzheimer’s diseases and in Down’s syndrome [Basso et al. 2004, Kim et al. 2001, Krapfenbauer et al. 2003]. Prx III has been found to protect hippocampal neurons from excitotoxic injury in vivo [Hattori et al. 2003]. Our previous study suggested that manganese SOD (MnSOD), thioredoxin (Trx), thioredoxin reductase (TrxR) and the catalytic (GLCL-c) and regulatory (GLCL-r) subunits of glutamate cysteine ligase (g-glutamylcysteinesynthetase) play an important role in the pathogenesis of diffusely infiltrating astrocytomas [Haapasalo et al. 2003]. In that paper, we compared the expression of these enzymes in a large series of diffuse astrocytomas and in a small series of pilocytic astrocytomas. In the present study, we principally investigate the expression of Prx I – VI in pilocytic astrocytomas in central nervous system. Pilocytic astrocytomas are one of the most common tumors in children, and in contrast to diffuse supratentorial astrocytomas in adults, they are often located in the cerebellum. They also have a totally different molecular genetic background. Our principal aim was to study the biology of peroxiredoxins in pilocytic astrocytomas, and our hypothesis was that they have a role in the pathogenesis of benign tumors. This has not been previously thoroughly examined. In addition, pilocytic astrocytomas offer a model without the bias produced by radiation or chemotherapy. We evaluated Prxs in association with age, survival, histopathological and molecular pathological features, including other antioxidant enzymes, in a series of 105 pilocytic astrocytomas.

Methods Immunohistochemistry of Prxs and other AOEs The immunostaining procedure was as follows. 4 m thick sections were cut from the microarray blocks, which were then deparaffinized in xylene and rehydrated in a descending ethanol series. In order to enhance immunoreactivity, the sections were incubated in 10 mM citrate buffer (pH 6.0), boiled in a microwave oven for 2 min at 850 W, and after that for 8 min at 350 W. Endogenous peroxidase activity was eliminated by incubation in 0.1% hydrogen peroxide in absolute methanol for 10 min. The polyclonal anti-Prx-antibodies were a gift from Dr. Kang (The Center for Cell Signalling Research and Division of Molecular Sciences, Ewha Womans University, Seoul, South Korea). Dilutions of the primary antibodies were 1 : 1,500 for Prx I, 1 : 1,000 for Prx II, 1 : 500 for Prx III, 1 : 1,000 for Prx IV and 1 : 2,000 for Prxs V and VI. Antibodies against other AOEs were as follows: polyclonal rabbit anti-human antibody to MnSOD (a gift from Professor J.D. Crapo, National Jewish Medical Center, Denver, CO, USA, dilution 1 : 1,000), rabbit polyclonal anti-human antibodies to GLCL-c and GLCL-r (a gift from Dr. Kavanagh, University of Washington, Seattle, WA, USA, dilution 1 : 1,000 for both), an affinity-purified goat-polyclonal human Trx antibody (American Diagnostica, Greenwich, CT, USA, dilution 1 : 200) and antibody to TrxR (a gift from Dr. Arne Holmgren, Karolinska Institutet, Stockholm, Sweden, dilution 1 : 1,000), which was the gammaglobulin fraction of a polyclonal rabbit anti-rat antibody directed against cytosolic TrxR in rat liver [Mustacich and Powis 2000, Nakamura et al. 1996]. Immunostaining of Prxs I – VI, MnSOD, GLCL-c, GLCL-r and TrxR was carried out using Histostain-Plus Kits (Zymed Laboratories Inc., South San Francisco, CA, USA) and the chromogen was aminoethyl carbazole (AEC) (Zymed Laboratories Inc.). For Trx, a biotinylated secondary anti-goat antibody was applied, followed by avidin-biotin peroxidase complex (both from Dakopatts, Glostrup, Denmark). Color was developed using 3,3’-diaminobenzidine, and the sections were lightly counterstained with hematoxylin and

NP-6036 / 22.03.2007

3

Peroxiredoxins in pilocytic astrocytomas

mounted in Eukitt (Kindler, Freiburg, Germany). Replacement of the primary antibody with phosphate-buffered saline (PBS) at pH 7.2, or with goat IgG immunoglobulin isotype (Zymed Laboratories, Inc., San Francisco, CA, USA) was carried for negative controls. The intensity of the immunostainings with all the antibodies was evaluated by dividing the staining reaction in 3 groups: 1 = weak staining intensity, 2 = moderate staining intensity, 3 = strong staining intensity. The quantity of the immunostaining was evaluated as follows: 0 = no positive immunostaining, 1 = < 25% of tumor cells showing positivity, 2 = 25 – 50% of tumor cells showing positivity, 3 = > 50% of tumor cells showing positivity. A combined score for the immunostaining, based on both qualitative and quantitative immunostaining was composed by adding both the qualitative and quantitative score which was then divided in 4 main groups: – = no immunostaining; score 0, + = weak immunostaining; scores 1 – 2, ++ = moderate immunostaining, scores 3 – 4, +++ = strong immunostaining, scores 5 – 6.

Proliferation index, p53 and apoptosis Ki-67MIB-1 and p53 were immunostained by using a standard streptavidin-biotin immunoperoxidase technique and scored as previously described [Haapasalo et al. 1999]. Apoptosis was detected by using ApopTagTM In Situ Apoptosis Detection Kits (Oncor Inc., Gaithersburg, MD, USA) as described previously [Haapasalo et al. 1999]. The microscope-based image analysis system (CAS200 Software, Becton Dickinson and Co., San Jose, CA, USA), equipped with 2 cameras for images of immunopositive (brown) and immunonegative (green) areas in nuclei, computed the proliferation indices from whole sections and the Ki-67MIB-1 labelling index (LI) [Haapasalo et al. 1999, Sallinen et al. 1994]. Tissues with unequivocal p53 staining of neoplastic nuclei (> 1/10 high power fields) were regarded as immunopositive [Haapasalo et al. 1993]. Apoptotic index was scored as percentages of ApopTag-immunoreactive cancer cell nuclei [Haapasalo et al. 1999].

Statistical analysis SPSS for Windows software (Chicago, IL, USA) was used for statistical analysis. The significance of associations was determined using Chi-square and Mann-Whitney tests. Kaplan-Meier curves and the log rank test were used in survival analysis.

Material The tumor specimens had been fixed in 4% phosphate-buffered formaldehyde, after which they had been processed into paraffin blocks. First, all HE-stained slides of the tumors were evaluated by two neuropathologists (HH together with HK, AP or LP), and histopathological typing and grading were carried out according to WHO criteria [WHO 2000]. Two neuropathologists (HH and HK) evaluated the presence or absence of the major histological features of pilocytic astrocytomas: atypia, necrosis, endothelial proliferation, vascular hyalinization, perivascular lymphocytes, cystic pattern, granular bodies and calcification. One neuropathologist (HH) pinpointed one histologically representative tumor region in each pilocytic astrocytoma. From this tumor region a sample was included in multi-tissue microarray blocks representing 105 astrocytic tumors [Kononen et al. 1998]. The microarray blocks were constructed with a custom-built instrument (Beecher Instruments, Silver Spring, MD, USA). The diameter of the tissue core in the microarray block was 600 mm.

Patients Brain tumor samples were obtained from 105 patients (49 females and 56 males) operated on because of pilocytic astrocytomas at the University Hospitals of Tampere, Helsinki, Kuopio and Turku, Finland, during 1980 – 1999. Age varied from newborn to 66 (median = 10, Mean ± SD = 15 ± 15) years. None of the patients received preoperative radiation and/or chemotherapy. The topography of the tumors was as follows: 85% were in the cerebellum, 5% in the cerebrum, 3% in the brainstem and spinal cord, and 7% in cra-

NP-6036 / 22.03.2007

4

Nordfors, Haapasalo, Helén et al.

Table 1. Peroxiredoxin expression in pilocytic astrocytomas (96 primaries and 9 residives). 0 = negative, 1,2,3 = weak, moderate and strong immunopositive, respectively (p = n.s., c2 test). Prx I Expression

Prx II

Prx III

Prx IV

Prx V

Prx VI

Prim

Resid

Prim

Resid

Prim

Resid

Prim

Resid

Prim

Resid

Prim

Resid

0

1

0

0

0

0

0

7

0

12

0

1

1

1

2

1

3

0

26

2

29

1

35

1

14

2

2

9

2

20

1

47

1

45

5

20

3

36

2

3

81

6

70

8

20

6

10

3

25

5

43

4

Figure 1. A: Prx I staining showing moderate cytoplasmic positivity in pilocytic astrocytoma with microcystic degeneration (× 630). B: Strong cytoplasmic Prx II positivity in densely packed astrocytic cells (× 630). C: Strong, dot-like Prx III cytoplasmic immunopositivity probably due to the mitochondrial location (× 630). D: MnSOD staining showing strong cytoplasmic immunopositivity (× 630).

nial nerves (mostly the optic nerve). There were 96 patients with primary tumors available and 9 patients with recurrences. Follow-up time of primary tumors varied from 0.5 – 19.5 (Mean ± SD = 7.0 ± 4.1) years.

Results The distribution of immunostaining (no immunostaining (0), weakly (1), moderately (2), strongly (3) positive) of Prxs in 96 primary and 9 recurrent tumors is shown in Table 1. In an area adjacent to tumor moderate expression of MnSOD was seen. Also weak

positivity for Prx II was observed while other AOEs were negative. In tumor cells the immunostaining was cytoplasmic, but Prxs I, II, IV, V and VI also showed variable nuclear positivity. Most (98%) of all tumors expressed positivity for Prx I (84% strongly, 11% moderately and 3% weakly). All tumors showed positivity for Prx II (4% strongly, 20% moderately and 76% weakly) and also for Prx III (26% strongly, 47% moderately, 27% weakly). For Prx IV, 92% of cases were positive (13% strongly, 49% moderately, 30% weakly) and 87% were positive for Prx V (29% strongly, 23% moderately and 35% weakly). Strong Prx VI expression was found in 45%, moder-

NP-6036 / 22.03.2007

5

Peroxiredoxins in pilocytic astrocytomas

Figure 2. The distribution of Prx immunostaining in 105 pilocytic astrocytomas. 0 = no immunostaining present; 1 = weak immunostaining; 2 = moderate immunostaining; 3 = strong immunostaining.

Figure 3. Kaplan-Meier curve showing recurrencefree survival in relation to Prx VI immunostaining in pilocytic astrocytomas (p = 0.032, log-rank test).

ate expression in 37% and weak expression in 15% of all tumors. The immunoreactivity patterns of Prxs in pilocytic astrocytomas are shown in Figure 1A,B,C and in Figure 2. An example of MnSOD immunoreactivity in a pilocytic astrocytoma is shown in Figure 1D. The expression patterns of Prxs, considered in the following analyses as either negative (no and weak immunostaining) or positive (moderate and strong immunopositivity) were next correlated with each other. When compared with each other there were associations between Prx I expression and that of Prx II (p < 0.001), Prx IV (p = 0.010) and Prx VI (p < 0.001, c2 test). In addition to Prx I, Prx II

was associated with Prx IV (p = 0.002) and Prx VI (p < 0.001). Similarly, Prx III correlated with Prx IV (p = 0.004). Prx IV correlated with all other Prxs (Prx V, p = 0.007; Prx VI, p = 0.010). Expression of Prx II, III, IV, GLCL-c and TrxR was associated with vascular pathology (endothelial proliferation, vascular hyalinization and perivascular lymphocytes), whereas Prx I, II and TrxR correlated with degenerative features (cystic pattern, granular bodies, calcification) (Table 2). Expression of GLCL-r and TrxR correlated significantly with an aggressive tumor growth pattern (atypia and necrosis) (Table 2). Proliferation by MIB-1 was greater in Prx VI- and TrxR-positive tumors (Prx VI: negative tumors, 1.8 ± 3.1 (Mean ± SD), positive tumors 3.0 ± 3.1, p = 0.037, TrxR: negative tumors, 2.2 ± 3.1, positive tumors, 3.5 ± 3.0, p = 0.028, Mann-Whitney test). However, MnSOD positivity was associated with lower proliferative activity (negative tumors, 3.3 ± 3.2, positive tumors 2.2 ± 2.9, p = 0.039, Mann-Whitney test). There was no correlation between AOE status and p53 immunopositivity or TUNEL (apoptosis) positivity (Mann-Whitney test). When clinical features were compared with the expression of Prx and other AOEs, MnSOD and GLCL-c positivity increased significantly with increasing patient age (p = 0.022 and p = 0.010, respectively, MannWhitney test). In addition, the expression of MnSOD was significantly lower in recurrences (p = 0.019, Mann-Whitney test). No such differences were seen as regards Prxs. Additionally, there was no association between the location of the tumors and the enzyme expression (c2 test). All 96 patients with primary pilocytic astrocytomas were included in survival analysis. Infrequent enzyme negativity of Prx I and II did not allow the possibility of survival analysis as regards these enzymes. Tumors immunopositive for Prx VI seemed to be associated with significantly better recurrence-free survival when compared with immunonegative tumors (p = 0.032, log-rank test) (Figure 3). However, when patient age, Ki-67 (MIB-1), location (cerebellum versus other location) and Prx VI immunopositivity were included to multivariate analysis, none of these factors predicted independently recurrence-free sur-

NP-6036 / 22.03.2007

6

Nordfors, Haapasalo, Helén et al.

Table 2. Peroxiredoxins and other antioxidant enzymes by histology in pilocytic astrocytomas (N=65, p = p-value, c2). neg = enzyme-negative; pos = enzyme-positive.

Prx I neg

pos



0

5

+

1

59



1

60

+

0

4

Prx II p

neg

pos

0

5

2

58

2

59

0

4

2

20

0

43

1

41

1

22

2

53

0

10

1

2

1

61

0

2

1

22

1

53

1

10

Prx III p

neg pos

Prx IV p

neg

pos

3

2

26

33

28

32

1

3

12

9

17

26

19

23

10

12

28

26

1

9

2

1

27

34

0

1

13

10

23

30

6

5

Prx V p

neg

pos

34

13

13

4

45

15

2

2

30

10

17

7

38

14

9

3

43

15

4

2

4

4

43

13

6

3

11

4

42

15

5

2

Prx V p

neg pos

p

atypia

n.s.

n.s.

2

3

16

44

18

43

0

4

10

12

8

35

10

32

8

15

16

39

2

8

1

2

17

45

1

1

10

13

13

41

5

6

n.s.

n.s.

n.s.

2

3

10

51

11

51

1

3

4

18

8

36

10

33

2

21

12

44

0

10

1

2

11

52

0

2

5

19

11

44

1

10

n.s.

necrosis

n.s.

n.s.

n.s.

n.s.

n.s.

n.s.

endothelial proliferation –

1

20

+

0

44

n.s.

0.046

0.023

n.s.

n.s.

n.s.

vascular hyalinization –

1

42

+

0

22

n.s.

n.s.

n.s.

n.s.

n.s.

n.s.

perivascular lymphocytes –

1

54

+

0

10



1

2

+

0

62



0

5

+

0

74



1

53

+

0

11

n.s.

n.s.

n.s.

0.015

n.s.

n.s.

cystic pattern

0.000

0.002

n.s.

n.s.

n.s.

n.s.

granular bodies

n.s.

n.s.

n.s.

n.s.

n.s.

n.s.

calcification

n.s.

n.s.

n.s.

vival (Cox multivariate analysis). In overall survival analysis none of the enzymes was of prognostic significance.

Discussion There is compelling evidence that ROS are linked to the pathogenesis and behavior of human malignancies [Kinnula and Crapo 2004, Kinnula et al. 2004]. A major hypothesis explaining the importance of oxidants and imbalance of the cellular redox state is an altered pro-oxidant intracellular environment that facilitates mutations and/or inactivation of tumor suppressor genes and activates on-

n.s.

n.s.

n.s.

cogenes. This leads to changes in cell growth and apoptosis, and finally the development of neoplasia [Burdon 1995, Osada and Takahashi 2002]. For example, it has been shown that the cellular redox state regulates several pathways important in carcinogenesis, including cMyc-, p53- and FAS-mediated apoptosis and ras-mediated epidermal growth factor (EGF) receptor-dependent angiogenesis [Casanova et al. 2002, Kasahara et al. 1997, Suhara et al. 1998]. It has also been shown that H2O2 produced in EGF-stimulated cells governs both the accumulation of PI 3,4,5triphosphate and activation of protein kinase Akt. It also appears that Prx II participates in growth factor signalling by removing H2O2

NP-6036 / 22.03.2007

7

Peroxiredoxins in pilocytic astrocytomas

[Kwon et al. 2004]. Several AOEs, including Prxs, also influence tumor resistance by inducing chemo- and radio resistance of tumor cells [Chung et al. 2001]. Normal tissue is protected against ROS by AOEs, including Prxs. We and others have shown that Prx proteins are expressed in neoplasms, e.g. in follicular thyroid carcinoma [Chung et al. 1999], malignant mesothelioma [Kinnula et al. 2002b], lung [Lehtonen et al. 2004] and breast cancer [Karihtala et al. 2003]. However, the expression of Prx enzymes has not previously been studied in pilocytic astrocytomas. These tumors usually have, if properly treated, a benign clinical course showing many histological features of degeneration/regression in addition to proliferative activity [Haapasalo et al. 1999], and in their treatment no radiation or chemotherapy is used. Consequently, influence of antioxidant enzymes on tumor behavior can be studied in these tumors without the bias produced by such treatments. According to our data, peroxiredoxin expression is a common phenomenon in pilocytic astrocytomas. Prx enzymes and other antioxidant enzymes are closely associated with tumor cell proliferation, immunopositive tumors usually having a greater proliferation rate. Although there was no association between apoptosis rate and AOEs, histopathologically degenerative and regressive features were often associated with Prx expression. The same was true for vascular pathology, e.g. endothelial proliferation as a sign of angiogenesis. Some AOEs were associated with histologically aggressive tumor features. Even though Prx VI was associated with higher cell proliferation, it seemed to be associated with significantly better recurrence-free survival. This might be due to association between Prx VI expression and young patient age. In the present study of pilocytic astrocytomas, Prxs and other AOEs were commonly expressed simultaneously. This suggests that the neoplastic tissue suffers from a constant oxidative load, leading to simultaneous upregulation of these enzymes. Co-expression and a relationship between Prxs have also been observed in previous studies of other neoplasms, indicating that such a phenomenon is common in tumors [Karihtala et al. 2003, Kinnula et al. 2002b]. An increased oxidative load may lead to degenerative features and cellular destruction in tumor tissue.

Some degenerative features, especially a cystic pattern, were closely associated with the expression Prx I, Prx II and TrxR. Cystic degeneration with Rosenthal fibres and calcification (often seen in pilocytic astrocytomas) implies cell death, which may be a sign of previous hypoxic periods. Elevated levels of AOEs in pilocytic astrocytomas with such degenerative features indicate the tendency of these tumors to suffer from oxidative stress and increased defence against ROS-mediated tissue damage. We also found a significant association between AOE expression (Prx II, III, IV, GLCL-c and TrxR) and vascular pathology, e.g. endothelial vascular proliferation. Hypoxia and ROS induce HIF-a, which stimulates angiogenic factors such as vascular endothelial growth factor (VEGF) [Kinnula and Crapo 2004]. In diffuse astrocytomas it is most probably hypoxia that induces the expression of VEGF in tumor cells. This binds to VEGF receptors (VEGFRs) in endothelial cells, leading to endothelial vascular proliferation. In pilocytic astrocytomas endothelial vascular proliferation is also a common feature and could be a consequence of ROS and AOE induction, but without such a sinister implication as in diffuse astrocytomas. In the present study, with 105 tumors, Prx VI and TrxR were significantly associated with higher proliferation indices. MnSOD, a putative tumor suppressor in the group of AOEs, has been widely investigated in malignant cells. Isolated overexpression of MnSOD decreases the growth, proliferation and invasion of cultured malignant cells [Bravard et al. 1992, Li et al. 1995, St Clair et al. 1992, Yan et al. 1996, Zhong et al. 1997] (reviewed by [Kinnula and Crapo 2004]). Interestingly, in the present study, MnSOD was overexpressed in those pilocytic astrocytomas that had a significantly decreased proliferation rate. This is in line with its putative tumor suppressor function, which is further supported by the fact that in the recurrences the expression of MnSOD was significantly decreased. In contrast to this, in our previous study, MnSOD positivity was associated with a significantly higher proliferation rate in 380 diffusely infiltrating astrocytomas [Haapasalo et al. 2003]. In that study, we concluded that down-regulation of MnSOD might be an event in the progression of diffusely infiltrat-

NP-6036 / 22.03.2007

8

Nordfors, Haapasalo, Helén et al.

ing astrocytomas. This might be a result of the loss of chromosome region 6q, the locus of the MnSOD gene, which frequently takes place in diffuse astrocytomas [Sallinen et al. 1997]. Such genetic changes do not occur in pilocytic astrocytomas, which generally have either a normal karyotype or only a few major cytogenetic aberrations. Many AOEs, such as MnSOD and Prxs, increase chemo- and radio resistance of tumor cells. Thus, in this context, therapy may also influence tumor behavior. Pilocytic astrocytomas are treated by surgery alone, while diffuse astrocytomas are also treated by means of radiotherapy and occasionally by means of chemotherapy. In diffuse astrocytomas the expression of AOEs may thus, in the late progression of the disease, induce more aggressive behavior. This phenomenon could also partly explain the fact that diffuse astrocytomas expressing MnSOD or Trx are associated with worse prognosis than pilocytic astrocytomas expressing the same molecules. In contrast, Prx VI was linked to significantly better recurrence-free survival in pilocytic astrocytomas. This is probably a result of the fact that Prx VI overexpression in pilocytic astrocytomas without the bias produced by radio- or chemotherapy decreases ROS activity and in this way might abrogate genomic instability and tumor progression. Prx VI could be used as a prognostic marker when planning therapy or follow-up for patients with pilocytic astrocytomas. Previously, only a few factors have been shown to be of prognostic significance in this disease [Haapasalo et al. 1999]. In earlier studies univariate statistical analyses revealed that partial resection, older patient age and histology [Haapasalo et al. 1999], especially pilomyxoid tumor variant [Fernandez et al. 2003] were associated with a worse prognosis. Overall, the role of antioxidant enzymes especially MnSOD and peroxiredoxins in human tumors is poorly understood and partly controversial. Our study suggests that several AOEs and Prxs, at least Prx VI probably play a role in tumor growth/invasion and, thus, in prognostication. However, an immunohistochemical study cannot give any suggestion about the functional activity or possible overoxidation of these proteins. In addition, the prognostic value of Prx VI needs to be evaluated with a large number of recurrent pilocytic

astrocytomas, perhaps with more thorough MRI and clinical analysis to differentiate the recurrent and residual tumors.

Conclusion In conclusion, our study shows that peroxiredoxins and other antioxidant enzymes are expressed in pilocytic astrocytomas and may modulate their behavior. Co-expression of several antioxidant enzymes was common in these tumors. Antioxidant enzymes had a connection with angiogenesis and cell proliferation. They were usually associated with an elevated proliferation rate, but the putative tumor suppressor MnSOD depressed proliferative activity. Most interestingly, degeneration and regressive features were associated with AOEs but seemed not to be associated with p53-mediated apoptotic activity. Elevated levels of AOEs in pilocytic astrocytomas with degenerative features indicate the tendency of these tumors to suffer from oxidative stress and increased defence against ROS-mediated tissue damage.

Acknowledgments We thank Mrs Reija Randen, Mrs Riitta Koivisto, Mr Jorma Lampinen, Mrs Päivi Koukkula and Mr Manu Tuovinen for skillful technical assistance. This work was supported by grants from the Cancer Society of Finland and the Medical Research fund of Tampere University Hospital.

References Basso M, Giraudo S, Corpillo D, Bergamasco B, Lopiano L, Fasano M. Proteome analysis of human substantia nigra in Parkinson’s disease. Proteomics. 2004; 4: 3943-5392. Bravard A, Sabatier L, Hoffschir F, Ricoul M, Luccioni C, Dutrillaux B. SOD2: a new type of tumor-suppressor gene? Int J Cancer. 1992; 51: 476-480. Burdon RH. Superoxide and hydrogen peroxide in relation to mammalian cell proliferation. Free Radic Biol Med. 1995; 18: 775-794. Casanova ML, Larcher F, Casanova B, Murillas R, Fernandez-Acenero MJ, Villanueva C, MartinezPalacio J, Ullrich A, Conti CJ, Jorcano JL. A critical role for ras-mediated, epidermal growth factor receptor-dependent angiogenesis in mouse skin carcinogenesis. Cancer Res. 2002; 62: 3402-3407.

NP-6036 / 22.03.2007

9

Peroxiredoxins in pilocytic astrocytomas Chung YM, Yoo YD, Park JK, Kim YT, Kim HJ. Increased expression of peroxiredoxin II confers resistance to cisplatin. Anticancer Res. 2001; 21: 1129-1133. Chung YM, Yoo YD, Park JK, Kim YT, Kim HJ. Peroxiredoxin I expression in human thyroid tumors. Cancer Lett. 1999; 145: 127-132. Fernandez C, Figarella-Branger D, Girard N, BouvierLabit C, Gouvernet J, Paz Paredes A, Lena G. Pilocytic astrocytomas in children: prognostic factors – a retrospective study of 80 cases. Neurosurgery. 2003; 53: 544-553. Fujii J, Ikeda Y. Advances in our understanding of peroxiredoxin, a multifunctional, mammalian redox protein. Redox Rep. 2002; 7: 123-130. Haapasalo H, Isola J, Sallinen P, Kalimo H, Helin H, Rantala I. Aberrant p53 expression in astrocytic neoplasms of the brain: association with proliferation. Am J Pathol. 1993; 142: 1347-1351. Haapasalo H, Kylaniemi M, Paunu N, Kinnula VL, Soini Y. Expression of antioxidant enzymes in astrocytic brain tumors. Brain Pathol. 2003; 13: 155-164. Haapasalo H, Sallinen S, Sallinen P, Helen P, Jaaskelainen J, Salmi TT, Paetau A, Paljarvi L, Visakorpi T, Kalimo H. Clinicopathological correlation of cell proliferation, apoptosis and p53 in cerebellar pilocytic astrocytomas. Neuropathol Appl Neurobiol. 1999; 25: 134-142. Hattori F, Murayama N, Noshita T, Oikawa S. Mitochondrial peroxiredoxin-3 protects hippocampal neurons from excitotoxic injury in vivo. J Neurochem. 2003; 86: 860-868. Karihtala P, Mantyniemi A, Kang SW, Kinnula VL, Soini Y. Peroxiredoxins in breast carcinoma. Clin Cancer Res. 2003; 9: 3418-3424. Kasahara Y, Iwai K, Yachie A, Ohta K, Konno A, Seki H, Miyawaki T, Taniguchi N. Involvement of reactive oxygen intermediates in spontaneous and CD95 (Fas/APO1)-mediated apoptosis of neutrophils. Blood. 1997; 89: 1748-1753. Kim SH, Fountoulakis M, Cairns N, Lubec G. Pro tein levels of human peroxiredoxin subtypes in brains of patients with Alz hei mer’s dis ease and Down syndrome. J Neural Transm. 2001; 61 (Suppl): 223- 235. Kinnula VL, Crapo JD. Superoxide dismutases in malignant cells and human tumors. Free Radic Biol Med. 2004; 36: 718-744. Kinnula VL, Lehtonen S, Kaarteenaho-Wiik R, Lakari E, Paakko P, Kang SW, Rhee SG, Soini Y. Cell-specific expression of peroxiredoxins in human lung and pulmonary sarcoidosis. Thorax. 2002a; 57: 157-164. Kinnula VL, Lehtonen S, Sormunen R, Kaarteenaho-Wiik R, Kang SW, Rhee SG, Soini Y. Overexpression of peroxiredoxins I, II, III, V and VI in malignant mesothelioma. J Pathol. 2002b; 196: 316-323. Kinnula VL, Pääkko P, Soini Y. Antioxidant enzymes and redox-regulating thiol proteins in malignancies of human lung. FEBS Lett. 2004; 569: 1-6. Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998; 4: 844-847. Krapfenbauer K, Engidawork E, Cairns N, Fountoulakis M, Lubec G. Aberrant expression of peroxiredoxin subtypes in neurodegenerative disorders. Brain Res. 2003; 967: 152-160. Kwon J, Lee SR, Yang KS, Ahn Y, Kim YJ, Stadtman ER, Rhee SG. Reversible oxidation and inactivation of the

tumor suppressor PTEN in cells stimulated with peptide growth factors. Proc Natl Acad Sci. 2004; 101: 16419-16424. Lehtonen ST, Svensk AM, Soini Y, Paakko P, Hirvikoski P, Kang SW, Saily M, Kinnula VL. Peroxiredoxins, a novel protein family in lung cancer. Int J Cancer. 2004; 111: 514-521. Li JJ, Oberley LW, St Clair DK, Ridnour LA, Oberley TD. Phenotypic changes induced in human breast cancer cells by overexpression of manganese-containing superoxide dismutase. Oncogene. 1995; 10: 1989-2000. Mustacich D, Powis G. Thioredoxin reductase. Biochem J. 2000; 346: 1-8. Nakamura M, Watanabe T, Klangby U, Asker C, Winman K, Yonekawa Y, Kleinhues P, Ohgaki O’Brien ML, Tew KD. Glutathione and related enzymes in multidrug resistance. Eur J Cancer. 1996; 32: 967-978. Osada H, Takahashi T. Genetic alterations of multiple tumor suppressors and oncogenes in the carcinogenesis and progression of lung cancer. Oncogene. 2002; 21: 7421-7434. Sallinen PK, Haapasalo HK, Visakorpi T, Helen PT, Rantala IS, Isola JJ, Helin HJ. Relation of Ki-67 (MIB-1), PCNA and S-phase fraction with patient survival in formalin-fixed, paraffin-embedded astrocytoma material. J Pathol. 1994; 174: 275-282. Sallinen SL, Sallinen P, Haapasalo H, Kononen J, Karhu R, Helen P, Isola J. Accumulation of genetic changes is associated with poor prognosis in Grade II astrocytomas. Am J Pathol. 1997; 151: 1799-1807. Sarafian TA, Verity MA, Vinters HV, Shih CC, Shi L, Ji XD, Dong L, Shau H. Differential expression of peroxiredoxin subtypes in human brain cell types. J Neurosci Res. 1999; 56: 206-212. St Clair DK, Wan XS, Oberley TD, Muse KE, St Clair WH. Suppression of radiation-induced neoplastic transformation by overexpression of mitochondrial superoxide dismutase. Mol Carcinog. 1992; 6: 238-242. Suhara T, Fukuo K, Sugimoto T, Morimoto S, Nakahashi T, Hata S, Shimizu M, Ogihara T. Hydrogen peroxide induces upregulation of Fas in human endothelial cells. J Immunol. 1998; 160: 4042-4047. Yan T, Oberley LW, Zhong W, St Clair DK. Manganese-containing superoxide dismutase overexpression causes phenotypic reversion in SV40-transformed human lung fibroblasts. Cancer Res. 1996; 56: 2864-2871. Zhong W, Oberley LW, Oberley TD, St Clair DK. Suppression of the malignant phenotype of human glioma cells by overexpression of manganese superoxide dismutase. Oncogene. 1997; 14: 481-490.

NP-6036 / 22.03.2007

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

Open Access

RESEARCH ARTICLE

The tumour-associated carbonic anhydrases CA II, CA IX and CA XII in a group of medulloblastomas and supratentorial primitive neuroectodermal tumours: an association of CA IX with poor prognosis Research article

Kristiina Nordfors*1, Joonas Haapasalo1, Miikka Korja2,3, Anssi Niemelä1, Jukka Laine4, Anna-Kaisa Parkkila5, Silvia Pastorekova6, Jaromir Pastorek6, Abdul Waheed7, William S Sly7, Seppo Parkkila8 and Hannu Haapasalo1

Abstract Background: Medulloblastomas (MBs) and supratentorial primitive neuroectodermal tumours (PNETs) are the most common highly aggressive paediatric brain tumours. In spite of extensive research on these tumours, there are only few known biomarkers or therapeutic target proteins, and the prognosis of patients with these tumours remains poor. Our aim was to investigate whether carbonic anhydrases (CAs), enzymes commonly overexpressed in various tumours including glioblastomas and oligodendrogliomas, are present in MBs and PNETs, and whether their expression can be correlated with patient prognosis. Methods: We determined the expression of the tumour-associated carbonic anhydrases CA II, CA IX and CA XII in a series of MB/PNET specimens (n = 39) using immunohistochemistry. Results: Endothelial CA II, cytoplasmic CA II, CA IX and CA XII were expressed in 49%, 73%, 23% and 11% of the tumours, respectively. CA II was detected in the neovessel endothelium and the tumour cell cytoplasm. CA IX was mainly expressed in the tumour cells located in perinecrotic areas. CA XII showed the most homogenous distribution within the tumours. Importantly, CA IX expression predicted poor prognosis in both univariate (p = 0.041) and multivariate analyses (p = 0.016). Conclusions: We suggest that CA IX should be considered a potential prognostic and therapeutic target in MBs and PNETs. Background Medulloblastomas (MBs) and primitive neuroectodermal tumours (PNETs) are classified as embryonal tumours of the central nervous system (CNS) and histologically correspond to WHO grade IV [1]. One viewpoint postulates that these tumours show a common ontogeny, arising from related progenitor cells that have the potential for divergent neuroepithelial differentation. However, in recent years molecular genetic analyses have demonstrated different genetic profiles for these tumours [1]. It * Correspondence: [email protected] 1

Department of Pathology, Tampere University Hospital, Tampere, Finland

Full list of author information is available at the end of the article

has been proposed that MBs originate from the neoplastic transformation of granule cell precursors in the cerebellum via deregulation of molecular pathways involved in normal cerebellar development [2,3]. Correspondingly, PNETs arise in the cerebral hemispheres, brain stem or spinal cord. The neuroepithelial tumour cells of a PNET may be undifferentiated or poorly differentiated. In addition, the tumour cells may have aberrant differentiations, including neuronal, astrocytic and ependymal lines. MB is the most common childhood malignant tumour of the central nervous system and accounts for 12-25% of all paediatric CNS tumours. It is very rare in adults, accounting for only 0.5-1% of brain tumours [4]. The

© 2010 Nordfors et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons

BioMed Central Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

main defective cell signalling pathways involved in the development of MB include the Hedgehog and Wnt pathways, but the exact molecular mechanisms contributing to tumourigenesis in both MB and PNET are still poorly understood [5]. MBs are sensitive to chemotherapy and radiation, but surgical resection continues to be the most effective treatment [6,7]. Patients with PNET undergo a similar treatment process to patients with MB [8]. There has been a marked improvement in the 5-year survival rate in MB patients, as the survival percentage has improved from 2-30% in the 1970s to 60-70% currently [1,4]. Unfortunately, the current clinical staging does not effectively identify the patients whose tumours will be resistant to chemotherapy and radiation. To individualise therapies and minimise side-effects of aggressive treatments, we need to overcome the major challenge of identifying the high- and low-risk patients. While the prognosis for patients with MB has improved, children with PNET have an even worse prognosis than patients with MB. Currently, the 5-year survival rate for patients with PNET is 24-38% [9,10]. The carbonic anhydrases (CAs) are zinc-containing metalloenzymes that catalyse the reversible hydration of carbon dioxide (CO2 + H2O ? HCO3- + H+), and, thus, participate in the maintenance of pH homeostasis in the body [11-14]. The mammalian α-CA gene family encodes at least thirteen enzymatically active isoforms with different structural and catalytic properties, and twelve of these are expressed in human tissues [15]. CA isozymes II, IX and XII have been associated with neoplastic processes, and they are potential histological and prognostic biomarkers of certain tumours, including diffuse astrocytomas [12,16,17]. CA II is the most widely distributed member of the CA gene family, being present in virtually every human tissue and organ. It is catalytically one of the most efficient enzymes known [18]. It is present to some extent in malignant cells, and, interestingly, it has been recently shown to be ectopically expressed in the endothelial cells of tumour neovessels [17,19]. Transmembrane enzyme, CA IX, was first recognised as a novel tumour-associated antigen expressed in several types of human carcinomas as well as in normal gastrointestinal tissue [12,20,21]. It has been functionally linked to cell adhesion, differentiation, proliferation and oncogenic processes [12,22], and its enzymatic activity is comparable to CA II [23]. Another transmembrane CA isozyme, CA XII, was first found in normal kidney tissue and renal cell carcinoma [24,25]. Later studies have shown that it is expressed in several other tumours, but also in some normal organs such as the colon and uterus [26,27]. CA IX and XII seem to be regulated by similar mechanisms, as transcription of these isozymes is induced in

Page 2 of 10

tumours under hypoxic conditions through hypoxia inducible factor-1 alpha (HIF-1α)-mediated pathways [28]. Even though very little is known about the regulation of CA II expression, it is unlikely that HIF-1α is involved. High expression of CA II, IX and XII in certain tumours, such as astrocytomas and oligodendrogliomas [16,17,29,30], has suggested that these enzymes may functionally participate in the invasion process, which is facilitated by acidification of the extracellular space [31]. In favour of this hypothesis, it has been shown in vitro that CA inhibitors can reduce the invasion capacity and proliferation of cancer cells [32-34]. To our knowledge, this is the first study to assess the expression of tumour-associated CAs in MBs and PNETs. Here we evaluate the expression of CA II, IX and XII in association with the patient age, survival and molecular pathologic features such as apoptosis and expression of cerbB2, MIB-1 and bcl-2.

Methods Study material

Brain tumour samples were obtained from 35 patients (15 females and 20 males) with either MBs or supratentorial PNETs who were operated on at the University Hospitals of Tampere and Turku, Finland, from1989-2005. The term supratentorial PNET is used as a synonym for CNS PNET, not otherwise specified [1]. MBs were observed in 28 patients and supratentorial PNETs in 7 patients. In addition, there were four patients with a recurrent tumour (two MBs: recurrence after 8 and 29 months in the cerebellum; two supratentorial PNETs: recurrences after 9 and 71 months in the brain stem and left frontal lobe, respectively). Taken together, our material included 39 surgical tumour samples. The age of the patients varied from newborn to 68 years (median = 7.4, mean ± SD = 14.4 ± 17.2), Table 1. In the early nineties eight-drugs-in-one -protocol and later vincristine, lomustine and prednisolon were widely used also for MB and PNETs [35]. Later the treatment in older children (over three years of age) started with radiation therapy with weekly vincristin doses (craniospinal dose 36 Gy and total tumor dose 54-55 Gy), and after irradiation a chemotherapy protocol using cisplatin, CCNU and vincristine was applied [36]. The later protocol is still in use. Children under three years of age have been treated with multidrug chemotherapy protocols from Childrens Cancer Group (USA) or German HITSKK-group generally without radiation therapy. Of the 35 patients, 4 received preoperative chemotherapy and/or radiation therapy, and these four patients all had a recurrent tumour. The tumours were radically resected if possible, and most patients were also treated with postoperative chemo- and/or radiotherapy as follows: three patients received surgery only, five patients were

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

Page 3 of 10

Table 1: Patient characteristics in different tumour subtypes and the correlation between them. MB

PNET

All primary tumours

p-value

15.0 ± 17.3

11.9 ± 17.8

14.4 ± 17.2

0.343*

Females

10

5

15

Male

18

2

20

Surgery only

3

2

3

Surgery +radiation

3

1

4

Surgery+chemotherapy

3

0

5

Surgery+radiation+chemo

19

4

23

Age (mean, years) Sex

0.088**

Therapy

0.540**

* Mann-Whitney test ** chi-square test

post-operatively treated with chemotherapy, four underwent radiotherapy and twenty-three patients received both postoperative chemotherapy and radiotherapy, Table 1. The overall survival was known for 35 patients, and 17 patients were alive and 18 patients dead at the end of the follow-up period. The 5-year survival for our patients was 46% in the total tumour material, 39% in MBs and 71% in PNETs. All the material was gathered from surgical operations. For immunohistochemistry, the brain tumour specimens were fixed immediately in 4% phosphate-buffered formaldehyde and processed into paraffin blocks. Haematoxylin and eosin-stained slides of the tumours were evaluated by two experienced neuropathologists, and the histopathological typing and grading were carried out according to WHO criteria [1]. Following the typing and grading of the specimens, a neuropathologist (HH) pinpointed one histologically representative area from each tumour with a high cellular proliferation index (as assessed by Ki-67 (MIB-1) staining) [37], and this area was then inserted into a multitissue block. The blocks were constructed with a custom-built instrument (Beecher Instruments, Silver Spring, MD) and the diameter of the tissue cores was 2 mm. Immunohistochemistry

The monoclonal antibody M75, recognising the N-terminal domain of human CA IX, has been described previously [20,21]. The rabbit anti-human CA XII antiserum against the secretory form of CA XII has been characterised by Karhumaa et al. [27]. Rabbit antiserum against human CA II has also been produced and characterised previously [38]. Normal rabbit serum (NRS) was used for control staining.

Immunohistochemical staining for CA II, CA IX and CA XII were performed using an automated immunostaining system with the Power Vision+ Poly-HRP IHC Kit reagents (ImmunoVision Technologies, Burlingame, CA). Briefly, the sections were: (a) rinsed in a wash buffer; (b) treated with 3% H2O2 in ddH2O for 5 min and rinsed in a wash buffer; (c) blocked with the Universal IHC Blocking/Diluent for 30 min and rinsed in a wash buffer; (d) incubated for 30 min with the rabbit anti-human CA II serum, rabbit-anti human CA XII serum, monoclonal M75 antibody or NRS diluted 1:2000 (rabbit sera) or 1:1000 (M75) in Universal IHC Blocking/Diluent; (e) rinsed in a wash buffer for 5 min three times; (f ) incubated in Poly-HRP-conjugated anti-rabbit/mouse IgG for 30 min and rinsed in a wash buffer for 5 min three times; (g) incubated in a DAB (3,3' diaminobenzidine tetrahydrochloride) solution (one drop DAB solution A and one drop DAB solution B with 1 ml ddH2O) for 6 min; (h) rinsed with ddH2O; (i) treated with CuSO4 for 5 min to enhance the signal and (j) rinsed with ddH2O. All procedures were carried out at room temperature. The sections were finally examined and photographed with a Zeiss Axioskop 40 microscope (Carl Zeiss; Göttingen, Germany). The staining reactivities for CA II, CA IX and CA XII were scored from the multitissue- blocks on a scale from 0 to 3 as follows: 0, no reaction; 1, weak reaction (< 10% positive cells); 2, moderate reaction (10-30% positive cells); 3, strong reaction (>30% positive cells). Due to the sample size, the staining results were categorised into two groups: negative staining was considered as CA-negative and weak, moderate and strong staining were considered as CA-positive.

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

The section preparation, immunostaining and analysis of apoptosis (TUNEL-labelling) [39] and the expression of c-erbB-2, p53 [40] and bcl-2 [41] were done as previously described. Statistical analysis

All statistical analyses were performed using SPSS 15.0 for Windows (Chicago, IL). The significance of the associations was defined using the chi-square test, the MannWhitney test and the Kruskal-Wallis test. A log rank test, Kaplan-Meier curves and Cox multivariate regression analysis were used in the survival analysis. Ethics

The study design was approved by the Ethics committee of Tampere University Hospital and the National Authority for Medicolegal Affairs.

Results Immunohistochemical staining of CA II, CA IX and CA XII in tumour specimens is shown in Figure 1. CA II showed two distinct staining patterns: the endothelium of neovessels and the cytoplasm of MB/PNET cells. Of all tumours, 49% (n = 18, 12 MBs/6 PNETs) stained positively for CA II in the tumour endothelium (32% strong, 11% moderate and 6% weak staining). Positive cytoplasmic CA II staining in tumour cells was found in 73% (n = 27, 20 MBs/7 PNETs) of the cases (11% strong, 38% moderate and 24% weak staining). CA IX and CA XII were less frequently expressed than CA II in the tumour samples: 23% (n = 9, 8 MBs/1 PNET) of the tumours were positive for CA IX (3% strong, 13% moderate and 7% weak staining), and only 11% (n = 4, 3 MBs/1 PNET) of the tumours were positive for CA XII (3% strong, 3% moderate and 5% weak staining). The CA IX-specific antibody stained perinecrotic areas in most of the tumours in which necrosis was visible. The CA XII was more homogenously distributed than CA IX, consistent with the results obtained previously in other tumours such as ovarian tumours [42]. Since MBs/PNETs are rare tumours and the availability of the specimens was limited to 39, the positively stained tumours (scores 1-3) were pooled for most statistical analyses, including the studies on patient survival. There was no significant correlation in the co-expression of CA II, IX and XII in the subgroups of MBs and PNETs nor did we found a correlation in the group of all tumours (chi-square test). We also compared CA II, IX and XII expression with various clinical features and molecular markers (Table 2). The expression of the CAs did not correlate with proliferation (MIB-1), apoptosis (chi-square and Mann-Whitney test) or expression of bcl-2, p53 or cerbB-2 in any of the groups except for the correlation between c-erbB-2 and CA IX in PNETs (p = 0.047, chi-

Page 4 of 10

square test). Interestingly, CA XII-positive staining correlated with younger patient age (total material p < 0.001, MBs p < 0.001, chi-square test, Table 2). Added to this, CA IX positivity associated with female gender (total material p = 0.048, MBs p = 0.023, Table 2, chi-square test). There was no significant difference in the expression of CAs between the primary and recurrent tumours in any of the groups (chi-square test). Moreover, there were no correlation between the tumour type (MBs/ PNETs) and CA intensity (chi-square test). All 35 patients with primary MB/PNET were included in the survival analysis (Figure 2). The patients with a CA IX-positive MB/PNET had a worse prognosis than those who had a CA IX-negative tumour (all tumours p = 0.041, MBs p = 0.030, PNETs p = n.s.; log-rank test; Figure 2). We also found a correlation between survival and CA XII staining in patients with MB. The patients with CA XIIpositive tumours showed significantly worse prognosis (p = 0.010, log-rank test). There was no significant difference in survival time between the histological subgroups (p = 0.463, log-rank test). Of the prognostic indicators used for MBs in the current WHO classification, (2007) the following variables were included into the Cox multivariate survival analysis: patient age, MIB-1 proliferation index, apoptosis index and expression of p53, c-erbB-2 and bcl-2. In addition, the histopathological group (MB vs. supratentorial PNET), CA II, CA IX and CA XII were used in the analysis. These variables were grouped as presented in Table 2. In the Cox analysis, only expression of CA IX (odds ratio 4.31; 95% confidence interval (CI) 1.31 - 14.11; p = 0.016) and the apoptosis index (odds ratio 3.29; 95% CI 1.05 - 10.31, p = 0.041) were independent prognostic factors. The expression of either CA II and CA XII failed to show any significant association with survival.

Discussion In the present study, we demonstrate that several MBs and supratentorial PNETs express the CA isozymes CA II, CA IX and CA XII. According to the univariate survival analysis, expression of CA IX was found to be associated with poor prognosis. Most importantly, the Cox multivariate analysis, which included patient age, tumour cell proliferation, apoptosis rate and several other molecular factors, demonstrated that CA IX expression and apoptotic activity were the only independent prognostic factors. The expression of CA XII in the tumour cells was associated with patient age, as previously reported for the expression of CA XII in patients with diffuse astrocytomas [29]. These findings reflect the fact that patient age is a significant factor that contributes to carcinogenesis by several mechanisms and that tumour phenotypes are different depending on the age of the patient.

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

Page 5 of 10

Figure 1 Representative immunostaining of CA enzymes in MBs. Panel A shows no immunoreaction for CA IX, whereas the tumour in panel B is strongly positive. Panel C demonstrates CA XII-positive immunoreactivity in tumour cells. In panel D, CA II-positive immunostaining is confined to the endothelium of small blood vessels (arrows). All magnifications ×400.

The expression of CA II, CA IX and CA XII in the normal nervous system has been investigated in several previous studies. The localization of CA II is well documented in the normal human oligodendrocytes [43]. Based on our previous studies, CA IX is not present in the normal human brain [16]. RT-PCR analysis has shown very weak CA XII mRNA expression in the human brain [29], and in mouse, immunohistochemical staining has located CA XII to the choroid plexus [44]. CA IX has several functions in tumour progression. It has been proposed to have a capacity to modulate E-cadherin-mediated cell adhesion, thus leading to a more aggressive phenotype of malignant cells. In intercellular junctions, CA IX may be linked to the E-cadherin/βcatenin complex, because CA IX co-immunoprecipitated with β-catenin in cultured MDCK cells, a kidney cell line [45]. It is also noteworthy that β-catenin is mutated in some sporadic cases of MB [46]. The presence of CA IX

in the E-cadherin/β-catenin complex might contribute by an unknown mechanism to increased invasion and spread of tumour cells. Indeed, embryonal tumours differ from other brain tumours by their tendency to metastasise. In addition, our previous findings in diffusely infiltrating astrocytomas are also in line with the suggested role of CAs in the invasion process. Typically, CA IX-positive astrocytic tumours are highly invasive tumours with an extremely poor prognosis [16]. A recent study by Chiche et al. [47] provided clear evidence that both CA IX and CA XII are functionally involved in tumour growth. In vivo experiments showed that CA9 gene silencing alone led to a 40% reduction in xenograft tumour volume, and the silencing of both CA9 and CA12 resulted in an 85% reduction in tumour volume. In this study, CA IX was found in the perinecrotic areas of the tumours whenever necrosis was present. A similar hypoxia-associated pattern of CA IX expression has been

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

Page 6 of 10

Table 2: Association of endothelial and cytoplasmic CA II, CA IX and CA XII immunostaining with clinicopathologic variables in medulloblastomas (MB) and primitive neuroectodermal tumours (PNET). endothelial CA II positivity N Primary tumors

cytoplasmic CA II positivity N

CA IX positivity N

CA XII positivity N

All

MB

PNET

All

MB

PNET

All

MB

PNET

All

MB

PNET

33

27

6

33

27

6

35

28

7

34

27

7

< 3 years

5

2

3

5

3

2

1

1

0

4*

3+

1

> 3 years

12

10

2

18

15

3

7

6

1

0

0

0

-female

6

3

3

10

7

3

1**

0++

1

1

0

1

-male

11

9

2

13

11

2

7

7

0

3

3

0

Age

Gender

Localization -cerebellum

12

18

7

3

-cerebrum

5

5

1

1

Total material MIB-1

36

28

8

36

28

8

37

29

8

36

28

8

-below median

9

6

3

13

10

3

5

4

1

1

1

0

-above median

9

6

3

13

9

4

4

4

0

2

1

1

Apoptosis

37

29

8

37

29

8

39

30

9

38

29

9

-below median

8

5

3

15

11

4

5

5

0

1

1

0

-above median

10

7

3

12

9

3

4

3

1

3

2

1

Bcl-2

37

29

8

37

29

8

39

30

9

38

29

9

-negative

6

4

2

13

9

4

3

2

1

1

0

1

-positive

12

8

4

14

11

3

6

6

0

3

3

0

P53

37

29

8

37

29

8

39

30

9

38

29

9

-negative

15

6

1

23

9

1

7

4

0

3

0

0

-positive

3

6

5

4

11

6

2

4

1

1

3

1

ErbB2

37

29

8

37

29

8

39

30

9

38

29

9

-negative

8

10

5

11

10

5

6

7



0

2

1

-positive

10

2

1

16

2

2

3

1

1

4

1

0

The total number of tumours analysed in each category is in bold. * p < 0.001, chi-square test ** p = 0.048, chi-square test + p < 0.001, chi-square test ++ p = 0.023, chi-square test Ψ p = 0.047, chi-square test

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

previously detected in astrocytic tumours [16]. Hypoxia triggers architectural and phenotypic rearrangements of tumour tissue, resulting in the development of necrotic areas surrounded by zones of surviving hypoxic cells. Importantly, these cells often become the most aggressive tumour cells [48], in which CA IX expression is induced by HIF1-α-regulated pathway [28]. Because necrosis is an uncommon feature and is not considered to be a significant prognostic factor in MBs, the induction of CA IX in MBs/PNETs may also involve hypoxia-independent mechanisms. Similarly, in previous studies on gliomas, CA IX expression was seen in tumour cells located in close proximity to the blood vessels [49], and it has been shown that acidosis induces CA IX independently of pericellular hypoxia in glioblastoma cell lines [31]. Based on the previous studies, it has become clear that although hypoxia is the key factor for CA IX induction, there may be other important factors involved. Our text already pointed out that tumor cell acidosis seems to contribute to the expression level [31]. There are several studies where CA IX expression has been correlated to pimonidazole accumulation. The results have shown slightly conflicting results, which may reflect to biological variation between different tumor types and dynamics of tumor hypoxia. However, most results give support for the idea that CA IX follows the pattern of pimonidazole binding [50,51]. We have previously studied the expression of tumourassociated CAs in other types of brain tumours. Endothelial CA II was expressed in the neovessels of astrocytic tumours, in which it was associated with poor prognosis [17]. In addition, we have shown that both CA IX and CA XII are independent prognostic factors in glial tumours [16,29,30]. Based on these findings, CAs may play a central role in the pathogenesis of malignant brain tumours and may represent potential biomarkers for histopathological diagnosis of brain tumours. Although in the total tumour material CA II and CA XII did not reach statistical significance in for use as prognostic indicators, CA II had a similar trend to that of CA IX. Furthermore, CA XII showed a significant correlation with survival in MBs. As discussed above, these differences may be partly explained by regulatory mechanisms. It has been also shown that higher CA IX expression is associated with a more favourable overall survival in some tumours, such as in renal cell carcinoma (RCC) and in acute myeloid leukemia (AML). In RCC the CA IX induction is associated with VHL-mutation and not with hypoxia as in brain tumours [52]. In AML the association has been discussed to be involved with immune system and T-cell response [53]. Hypoxia-induced CA XII is less frequently expressed in MBs/PNETs than in gliomas. Interestingly, CA II was, once again, found in the endothelium of

Page 7 of 10

neovessels and, thus, may play an important functional role in tumour metabolism. In melanoma patients, endothelial CA II represents a major target antigen in dendritic cell therapy [19]. Further studies are, therefore, clearly warranted to evaluate the role of CA II as a possible therapeutic target not only in melanoma but also in other forms of cancer, including MBs/PNETs. CA IX-specific inhibitors would represent ideal candidate molecules for cancer therapy, because CA IX is highly expressed in several cancers while it shows a very limited distribution in normal tissues [22]. Design of isozyme-specific inhibitor has proved to be a great challenge, because the CA active site is quite similar in all active alpha CA isoforms. The recently published crystal structure of CA IX was certainly a major breakthrough that will help to design novel inhibitors with higher specificity [54]. According to our results, apoptosis was another independent prognostic factor in MBs/PNETs, although its role seems to be controversial. On one hand, as we show in this study, a higher apoptotic index is associated with better prognosis [55]. On the other hand, previous studies in which the degree of apoptosis was categorised as 'focal', 'diffuse' or 'extensive' demonstrated a correlation only between survival and focal apoptosis [6]. In addition, it is known that bcl-2 is an inhibitor of apoptosis. In our tissue samples, however, immunoreactivity for bcl-2 did not correlate with better prognosis; although similar results have been reported by others [56]. In our study, apoptosis was an independent prognostic indicator of MBs/PNETs. However, the study material was rather limited due to the fact that MBs and PNETs are rare tumours, and studies will be needed to clarify the association between apoptosis and survival. Added to this, the time period in which the patients were treated, was rather long and treatment protocols varied. In the future, children diagnosed with MB/PNET will be more accurately stratified based on a combination of clinical variables and molecular profiles. Improved risk stratification will enable individualised therapies, which could be a combination of conventional treatment modalities and novel, targeted therapeutic approaches. These changes will hopefully result in improved survival without a detriment in the quality of life. Several molecular alterations have already been identified in MBs, many of which appear to have prognostic significance.

Conclusions Based on our results, CA IX seems to be a promising prognostic marker that should be tested in a larger cohort of MB/PNET patients. The expression of CA IX in some MBs/PNETs suggests that it could be considered a potential therapeutic target, similar to other tumours including

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

Page 8 of 10

A

B

C

D

Figure 2 Kaplan-Meier curves showing overall survival of patients with MB or PNET categorised by: A. tumour cell-associated CA II, B. endothelial CA II, C. CA IX (p = 0.041; log-rank test), and D. CA XII immunostaining results.

acute myeloid leukemia [53] and renal cancer [57]. Furthermore, CA IX could be used for in vivo imaging and as a target molecule for CA inhibitors [12,22]. Competing interests The authors declare that they have no competing interests. Authors' contributions KN as the corresponding author gathered the clinical data of the patients, made the statistical analyses, and drafted the first version of the manuscript. JH also participated in the collection of the clinical information, helped with the statistical analyses, and contributed to the writing of the first version of the manuscript. HH was the main organizer of the study. MK provided the clinical knowledge needed and chose the patients for the study at the University Hospital of Turku. AN, JL and HH performed essential microscopic analyses. AP and SPar provided further knowledge on CAs and were in charge of the microscopic analyses on CAs in MBs and PNETs. SPas and JP provided the antibodies against human CA IX, and the antibodies against human CA XII were from the laboratory of AW and WSS. All authors read and approved the final manuscript. Acknowledgements We thank Mikko Arola, MD, PhD for his support in gathering detailed oncological treatment protocols. We also thank Aulikki Lehmus and Reija Randen for their skilful technical assistance. This work was supported by grants from the Cancer Society of Finland, EU 6th Framework programme (DeZnIT), Medical Research Fund of Tampere University Hospital, The Finnish Medical Foundation and The Nona and Kullervo Väre Foundation.

Author Details 1Department of Pathology, Tampere University Hospital, Tampere, Finland, 2Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland, 3Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland, 4Department of Pathology, Turku University Hospital, Turku, Finland, 5Department of Neurology, Tampere University Hospital, Tampere, Finland, 6Centre of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovak Republic, 7Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, MO, USA and 8Institute of Medical Technology and School of Medicine, University of Tampere and Tampere University Hospital, Tampere, Finland Received: 9 November 2009 Accepted: 18 April 2010 Published: 18 April 2010 © This BMC 2010 is article Cancer an Nordfors Open is2010, available Access et10:148 al; licensee from: articlehttp://www.biomedcentral.com/1471-2407/10/148 distributed BioMed Central underLtd. the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

References 1. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK: WHO Classification of Tumours of the Central Nervous System. IARC:Lyon; 2007:132-146. 2. Vandenberg SR, Herman MM, Rubinstein LJ: Embryonal central neuroepithelial tumors: current concepts and future challenges. Cancer Metastasis Rev 1987, 5(4):343-365. 3. Shakhova O, Leung C, van Montfort E, Berns A, Marino S: Lack of Rb and p53 delays cerebellar development and predisposes to large cell anaplastic medulloblastoma through amplification of N-Myc and Ptch2. Cancer Res 2006, 66(10):5190-5200. 4. Sarkar C, Deb P, Sharma MC: Medulloblastomas: new directions in risk stratification. Neurol India 2006, 54(1):16-23.

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

5.

6.

7.

8.

9.

10.

11. 12.

13. 14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

Fogarty MP, Kessler JD, Wechsler-Reya RJ: Morphing into cancer: the role of developmental signaling pathways in brain tumor formation. J Neurobiol 2005, 64(4):458-475. Giangaspero F, Wellek S, Masuoka J, Gessi M, Kleihues P, Ohgaki H: Stratification of medulloblastoma on the basis of histopathological grading. Acta Neuropathol 2006, 112(1):5-12. Rood BR, Macdonald TJ, Packer RJ: Current treatment of medulloblastoma: recent advances and future challenges. Semin Oncol 2004, 31(5):666-675. McBride SM, Daganzo SM, Banerjee A, Gupta N, Lamborn KR, Prados MD, Berger MS, Wara WM, Haas-Kogan DA: Radiation is an important component of multimodality therapy for pediatric non-pineal supratentorial primitive neuroectodermal tumors. Int J Radiat Oncol Biol Phys 2008, 72(5):1319-1323. Geyer JR, Sposto R, Jennings M, Boyett JM, Axtell RA, Breiger D, Broxson E, Donahue B, Finlay JL, Goldwein JW, et al.: Multiagent chemotherapy and deferred radiotherapy in infants with malignant brain tumors: a report from the Children's Cancer Group. J Clin Oncol 2005, 23(30):7621-7631. Pizer BL, Clifford SC: The potential impact of tumour biology on improved clinical practice for medulloblastoma: progress towards biologically driven clinical trials. Br J Neurosurg 2009, 23(4):364-375. Supuran CT: Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov 2008, 7(2):168-181. Pastorekova S, Parkkila S, Pastorek J, Supuran CT: Carbonic anhydrases: current state of the art, therapeutic applications and future prospects. J Enzyme Inhib Med Chem 2004, 19(3):199-229. Sly WS, Hu PY: Human carbonic anhydrases and carbonic anhydrase deficiencies. Annu Rev Biochem 1995, 64:375-401. Scozzafava A, Mastrolorenzo A, Supuran CT: Carbonic anhydrase inhibitors and activators and their use in therapy. Expert Opin Ther Patents 2006, 16(12):1627-1664. Hilvo M, Tolvanen M, Clark A, Shen B, Shah GN, Waheed A, Halmi P, Hanninen M, Hamalainen JM, Vihinen M, et al.: Characterization of CA XV, a new GPI-anchored form of carbonic anhydrase. Biochem J 2005, 392(Pt 1):83-92. Haapasalo JA, Nordfors KM, Hilvo M, Rantala IJ, Soini Y, Parkkila AK, Pastorekova S, Pastorek J, Parkkila SM, Haapasalo HK: Expression of carbonic anhydrase IX in astrocytic tumors predicts poor prognosis. Clin Cancer Res 2006, 12(2):473-477. Haapasalo J, Nordfors K, Jarvela S, Bragge H, Rantala I, Parkkila AK, Haapasalo H, Parkkila S: Carbonic anhydrase II in the endothelium of glial tumors: a potential target for therapy. Neuro Oncol 2007, 9(3):308-313. Kivela AJ, Kivela J, Saarnio J, Parkkila S: Carbonic anhydrases in normal gastrointestinal tract and gastrointestinal tumours. World J Gastroenterol 2005, 11(2):155-163. Yoshiura K, Nakaoka T, Nishishita T, Sato K, Yamamoto A, Shimada S, Saida T, Kawakami Y, Takahashi TA, Fukuda H, et al.: Carbonic anhydrase II is a tumor vessel endothelium-associated antigen targeted by dendritic cell therapy. Clin Cancer Res 2005, 11(22):8201-8207. Pastorekova S, Zavadova Z, Kostal M, Babusikova O, Zavada J: A novel quasi-viral agent, MaTu, is a two-component system. Virology 1992, 187(2):620-626. Chrastina A, Zavada J, Parkkila S, Kaluz S, Kaluzova M, Rajcani J, Pastorek J, Pastorekova S: Biodistribution and pharmacokinetics of 125I-labeled monoclonal antibody M75 specific for carbonic anhydrase IX, an intrinsic marker of hypoxia, in nude mice xenografted with human colorectal carcinoma. Int J Cancer 2003, 105(6):873-881. Pastorekova S, Parkkila S, Zavada J: Tumor-associated carbonic anhydrases and their clinical significance. Adv Clin Chem 2006, 42:167-216. Hilvo M, Baranauskiene L, Salzano AM, Scaloni A, Matulis D, Innocenti A, Scozzafava A, Monti SM, Di Fiore A, De Simone G, et al.: Biochemical characterization of CA IX, one of the most active carbonic anhydrase isozymes. J Biol Chem 2008, 283(41):27799-27809. Tureci O, Sahin U, Vollmar E, Siemer S, Gottert E, Seitz G, Parkkila AK, Shah GN, Grubb JH, Pfreundschuh M, et al.: Human carbonic anhydrase XII: cDNA cloning, expression, and chromosomal localization of a carbonic anhydrase gene that is overexpressed in some renal cell cancers. Proc Natl Acad Sci USA 1998, 95(13):7608-7613. Ivanov SV, Kuzmin I, Wei MH, Pack S, Geil L, Johnson BE, Stanbridge EJ, Lerman MI: Down-regulation of transmembrane carbonic anhydrases

Page 9 of 10

26.

27.

28.

29.

30.

31.

32.

33. 34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

in renal cell carcinoma cell lines by wild-type von Hippel-Lindau transgenes. Proc Natl Acad Sci USA 1998, 95(21):12596-12601. Kivela A, Parkkila S, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, Waheed A, Sly WS, Grubb JH, Shah G, et al.: Expression of a novel transmembrane carbonic anhydrase isozyme XII in normal human gut and colorectal tumors. Am J Pathol 2000, 156(2):577-584. Karhumaa P, Parkkila S, Tureci O, Waheed A, Grubb JH, Shah G, Parkkila A, Kaunisto K, Tapanainen J, Sly WS, et al.: Identification of carbonic anhydrase XII as the membrane isozyme expressed in the normal human endometrial epithelium. Mol Hum Reprod 2000, 6(1):68-74. Wykoff CC, Beasley NJ, Watson PH, Turner KJ, Pastorek J, Sibtain A, Wilson GD, Turley H, Talks KL, Maxwell PH, et al.: Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res 2000, 60(24):7075-7083. Haapasalo J, Hilvo M, Nordfors K, Haapasalo H, Parkkila S, Hyrskyluoto A, Rantala I, Waheed A, Sly WS, Pastorekova S, et al.: Identification of an alternatively spliced isoform of carbonic anhydrase XII in diffusely infiltrating astrocytic gliomas. Neuro Oncol 2008, 10(2):131-138. Jarvela S, Parkkila S, Bragge H, Kahkonen M, Parkkila AK, Soini Y, Pastorekova S, Pastorek J, Haapasalo H: Carbonic anhydrase IX in oligodendroglial brain tumors. BMC Cancer 2008, 8:1. Ihnatko R, Kubes M, Takacova M, Sedlakova O, Sedlak J, Pastorek J, Kopacek J, Pastorekova S: Extracellular acidosis elevates carbonic anhydrase IX in human glioblastoma cells via transcriptional modulation that does not depend on hypoxia. Int J Oncol 2006, 29(4):1025-1033. Parkkila S, Rajaniemi H, Parkkila AK, Kivela J, Waheed A, Pastorekova S, Pastorek J, Sly WS: Carbonic anhydrase inhibitor suppresses invasion of renal cancer cells in vitro. Proc Natl Acad Sci USA 2000, 97(5):2220-2224. Supuran CT: Indisulam: an anticancer sulfonamide in clinical development. Expert Opin Investig Drugs 2003, 12(2):283-287. Supuran CT, Briganti F, Tilli S, Chegwidden WR, Scozzafava A: Carbonic anhydrase inhibitors: sulfonamides as antitumor agents? Bioorg Med Chem 2001, 9(3):703-714. Finlay JL, Boyett JM, Yates AJ, Wisoff JH, Milstein JM, Geyer JR, Bertolone SJ, McGuire P, Cherlow JM, Tefft M, et al.: Randomized phase III trial in childhood high-grade astrocytoma comparing vincristine, lomustine, and prednisone with the eight-drugs-in-1-day regimen. Childrens Cancer Group. J Clin Oncol 1995, 13(1):112-123. Packer RJ, Sutton LN, Elterman R, Lange B, Goldwein J, Nicholson HS, Mulne L, Boyett J, D'Angio G, Wechsler-Jentzsch K, et al.: Outcome for children with medulloblastoma treated with radiation and cisplatin, CCNU, and vincristine chemotherapy. J Neurosurg 1994, 81(5):690-698. Sallinen PK, Haapasalo HK, Visakorpi T, Helen PT, Rantala IS, Isola JJ, Helin HJ: Prognostication of astrocytoma patient survival by Ki-67 (MIB-1), PCNA, and S-phase fraction using archival paraffin-embedded samples. J Pathol 1994, 174(4):275-282. Parkkila AK, Parkkila S, Juvonen T, Rajaniemi H: Carbonic anhydrase isoenzymes II and I are present in the zona glomerulosa cells of the human adrenal gland. Histochemistry 1993, 99(1):37-41. Miettinen HE, Jarvinen TA, Kellner U, Kauraniemi P, Parwaresch R, Rantala I, Kalimo H, Paljarvi L, Isola J, Haapasalo H: High topoisomerase IIalpha expression associates with high proliferation rate and and poor prognosis in oligodendrogliomas. Neuropathol Appl Neurobiol 2000, 26(6):504-512. Haapasalo H, Hyytinen E, Sallinen P, Helin H, Kallioniemi OP, Isola J: cerbB-2 in astrocytomas: infrequent overexpression by immunohistochemistry and absence of gene amplification by fluorescence in situ hybridization. Br J Cancer 1996, 73(5):620-623. Kallio JP, Hirvikoski P, Helin H, Luukkaala T, Tammela TL, KellokumpuLehtinen P, Martikainen PM: Renal cell carcinoma MIB-1, Bax and Bcl-2 expression and prognosis. J Urol 2004, 172(6 Pt 1):2158-2161. Hynninen P, Vaskivuo L, Saarnio J, Haapasalo H, Kivela J, Pastorekova S, Pastorek J, Waheed A, Sly WS, Puistola U, et al.: Expression of transmembrane carbonic anhydrases IX and XII in ovarian tumours. Histopathology 2006, 49(6):594-602. Kumpulainen T, Dahl D, Korhonen LK, Nystrom SH: Immunolabeling of carbonic anhydrase isoenzyme C and glial fibrillary acidic protein in paraffin-embedded tissue sections of human brain and retina. J Histochem Cytochem 1983, 31(7):879-886.

Nordfors et al. BMC Cancer 2010, 10:148 http://www.biomedcentral.com/1471-2407/10/148

44. Kallio H, Pastorekova S, Pastorek J, Waheed A, Sly WS, Mannisto S, Heikinheimo M, Parkkila S: Expression of carbonic anhydrases IX and XII during mouse embryonic development. BMC Dev Biol 2006, 6:22. 45. Svastova E, Zilka N, Zat'ovicova M, Gibadulinova A, Ciampor F, Pastorek J, Pastorekova S: Carbonic anhydrase IX reduces E-cadherin-mediated adhesion of MDCK cells via interaction with beta-catenin. Exp Cell Res 2003, 290(2):332-345. 46. Zurawel RH, Chiappa SA, Allen C, Raffel C: Sporadic medulloblastomas contain oncogenic beta-catenin mutations. Cancer Res 1998, 58(5):896-899. 47. Chiche J, Ilc K, Laferriere J, Trottier E, Dayan F, Mazure NM, Brahimi-Horn MC, Pouyssegur J: Hypoxia-inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH. Cancer Res 2009, 69(1):358-368. 48. Diaz-Gonzalez JA, Russell J, Rouzaut A, Gil-Bazo I, Montuenga L: Targeting hypoxia and angiogenesis through HIF-1alpha inhibition. Cancer Biol Ther 2005, 4(10):1055-1062. 49. Proescholdt MA, Mayer C, Kubitza M, Schubert T, Liao SY, Stanbridge EJ, Ivanov S, Oldfield EH, Brawanski A, Merrill MJ: Expression of hypoxiainducible carbonic anhydrases in brain tumors. Neuro Oncol 2005, 7(4):465-475. 50. Goethals L, Debucquoy A, Perneel C, Geboes K, Ectors N, De Schutter H, Penninckx F, McBride WH, Begg AC, Haustermans KM: Hypoxia in human colorectal adenocarcinoma: comparison between extrinsic and potential intrinsic hypoxia markers. Int J Radiat Oncol Biol Phys 2006, 65(1):246-254. 51. Shin KH, Diaz-Gonzalez JA, Russell J, Chen Q, Burgman P, Li XF, Ling CC: Detecting changes in tumor hypoxia with carbonic anhydrase IX and pimonidazole. Cancer Biol Ther 2007, 6(1):70-75. 52. Patard JJ, Fergelot P, Karakiewicz PI, Klatte T, Trinh QD, Rioux-Leclercq N, Said JW, Belldegrun AS, Pantuck AJ: Low CAIX expression and absence of VHL gene mutation are associated with tumor aggressiveness and poor survival of clear cell renal cell carcinoma. Int J Cancer 2008, 123(2):395-400. 53. Greiner J, Schmitt M, Li L, Giannopoulos K, Bosch K, Schmitt A, Dohner K, Schlenk RF, Pollack JR, Dohner H, et al.: Expression of tumor-associated antigens in acute myeloid leukemia: Implications for specific immunotherapeutic approaches. Blood 2006, 108(13):4109-4117. 54. Alterio V, Hilvo M, Di Fiore A, Supuran CT, Pan P, Parkkila S, Scaloni A, Pastorek J, Pastorekova S, Pedone C, et al.: Crystal structure of the catalytic domain of the tumor-associated human carbonic anhydrase IX. Proc Natl Acad Sci USA 2009, 106(38):16233-16238. 55. Haslam RH, Lamborn KR, Becker LE, Israel MA: Tumor cell apoptosis present at diagnosis may predict treatment outcome for patients with medulloblastoma. J Pediatr Hematol Oncol 1998, 20(6):520-527. 56. Nakasu S, Nakasu Y, Nioka H, Nakajima M, Handa J: bcl-2 protein expression in tumors of the central nervous system. Acta Neuropathol 1994, 88(6):520-526. 57. Davis ID, Liu Z, Saunders W, Lee FT, Spirkoska V, Hopkins W, Smyth FE, Chong G, Papenfuss AT, Chappell B, et al.: A pilot study of monoclonal antibody cG250 and low dose subcutaneous IL-2 in patients with advanced renal cell carcinoma. Cancer Immun 2007, 7:14. Pre-publication history The pre-publication history for this paper can be accessed here: http://www.biomedcentral.com/1471-2407/10/148/prepub doi: 10.1186/1471-2407-10-148 Cite this article as: Nordfors et al., The tumour-associated carbonic anhydrases CA II, CA IX and CA XII in a group of medulloblastomas and supratentorial primitive neuroectodermal tumours: an association of CA IX with poor prognosis BMC Cancer 2010, 10:148

Page 10 of 10