Overexpression in Childhood Brain Tumors - In Vivo

2 downloads 0 Views 194KB Size Report
primary childhood brain tumors was determined and the distribution pattern of ..... receptor may now be added to the long list of brain tumor progression markers and ... population-based study in Finland during 1975-93. Acta. Paediatrica 86: ...
in vivo 20: 519-526 (2006)

Cyclooxygenase-2 (COX-2) Overexpression in Childhood Brain Tumors BELA BODEY1,2, STUART E. SIEGEL2,3 and HANS E. KAISER3,4 1Department

of Pathology and 3Department of Pediatrics, University of Southern California, Keck School of Medicine, Los Angeles, CA; 2Childrens Center for Cancer and Blood Diseases, Childrens Hospital Los Angeles, Los Angeles, CA; 4Department of Pathology, School of Medicine, University of Maryland, Baltimore, MD, U.S.A. and Department of General and Experimental Pathology, University of Vienna, Vienna, Austria

Abstract. The overexpression of COX enzymes has been demonstrated in human neoplasms at various sites, including the colon, gastrointestinal tract, lung, skin and recently in brain tumors. In this study, COX-2 receptor overexpression in primary childhood brain tumors was determined and the distribution pattern of COX-2 receptors was examined. A sensitive, 4-step, alkaline phosphatase conjugated antigen detection technique was used and a specific monoclonal antibody for medulloblastomas/ primitive neuroectodermal tumors (MEDs/PNETs), anaplastic, high-grade astrocytomas (ASTRs) and in glioblastoma multiformes (GMs) was employed. All of the 14 MEDs/PNETs observed demonstrated high levels of immunoreactivity (overexpression), with the highest immunostaining intensity (grades A and B). However, of the 14 subtypes of astrocytic tumors examined, the COX-2 receptor expression level did not even approach those of the MEDs/PNETs levels. However, significant differences were found when comparing low grade pilocytic ASTRs to high grade anaplastic ASTRs and glioblastomas. In two low grade pilocytic ASTRs, the expression level never exceeded 20%, while in high grade glial tumors (6 anaplastic ASTRs and 6 GMs) 30 to 50% of the tumor cells overexpressed COX-2 receptors, documenting an increase in COX-2 receptor overexpression with the increasing grade of the astrocytic tumor. In view of these findings, it would appear likely that COX-2 inhibitors may represent a chemo-preventive tool in

Correspondence to: Bela Bodey, MD, D.Sc., 6820 Remmet Ave. Unit 201, Canoga Park, CA 91303, U.S.A. Tel: (818) 716-8612, email: [email protected] Key Words: Prostaglandin (PGE2) synthases or cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2), childhood medulloblastomas (MEDs)/primitive neuroectodermal tumors (PNETs), astrocytoma (ASTR), anaplastic astrocytoma (AA), glioblastoma multiforme (GBM), immunohistochemistry, immunophenotype (IP).

0258-851X/2006 $2.00+.40

treating childhood brain tumors, which are the leading cause of solid tumor cancer death in children under the age of 20. Both prostaglandin (PG) synthases, also called cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2), are catalytic enzymes involved in PG synthesis from arachidonic acid (1-4). Prostaglandin E2 (PGE2) functions promote the primary process of carcinogenesis and its further consolidation and progression via increased cell proliferation, decreased natural killer cell activity, in situ immune downmodulation, induction of neo-angiogenesis and the elevated expression of anti-apoptotic protein Bcl-2 (5). In addition, PGE2 significantly alters immune surveillance. COX-2, a principal COX isoenzyme, is induced in response to the actions of growth factors, various cytokines and promoters of neoplastic cell transformation and progression (4, 6, 7). An overexpression of COX enzymes has been demonstrated in human neoplasms at various sites, including the colon, gastrointestinal tract, lung, and skin (8). Immunohistochemical analyses of COX-2 expression identified that it is elevated in up to 90% of sporadic colon carcinomas and 40% of colonic adenomas (9, 10); it is also overexpressed in 40% to 80% of malignancies of the lung, head and neck, breast, prostate, brain and pancreas (11-13). Increased levels of COX-2, prostaglandins, or both, are found in adenomas in patients with familial adenomatous polyposis (14). Childhood brain tumors are now the leading cause of solid tumor cancer death in children under the age of 20, having surpassed acute lymphoblastic leukemia. The annual incidence of pediatric brain tumors appears to be on the rise, caused partly by improvements of diagnostic neuroimaging and its increased availability (15). Despite significant increases in the survival rate during the past decade, the great majority of pediatric patients with medulloblastoma (MED) and primitive neuroectodermal brain tumors (PNETs) still succumb to their disease (16-21). Historically, this common, childhood cerebellar malignancy was first described as MED by Bailey and Cushing

519

in vivo 20: 519-526 (2006) (22), based on the developmental theory of Schaper (23), who found apolar, "indifferent" cells in the external granular layer of the cerebellum and described them as the common neural stem cell or "medulloblast." Despite numerous cytological, histochemical, and electron microscopic studies, evidence for the real existence of a common neural stem cell, the hypothetical medulloblast, is still lacking. Morphological observations and comparative statistics over the past 90 years have failed to define cellular or brain tumor tissue features that have unequivocal prognostic value (22, 24-26). Hart and Earle introduced the concept of PNET to describe a series of supratentorial tumors that contained 95% or more of small undifferentiated cells not referable to the then current classification (27). No implication was made of a common neural stem cell origin, nor was there a hypothesis of developmental stage-dependent expression of antigens. Rorke (28, 29) called the PNET a "wastebasket category" and proposed a revised World Health Organi-zation (WHO) brain tumor classification based upon the intratumoral degree and type, as well as the main direction of cytomorphological differentiation (30). Rubinstein, however, expressed doubt about the common tissue origin and bracketing of infratentorial and supratentorial PNETs (sPNETs), such as MED, ependymoblastoma, central neuroblastoma, pineoblastoma (31), which are all referred to as PNETs according to the proposed classification (30). Morphologically, both MEDs and sPNETs are hypercellular and can be heterogeneous, containing areas of mixed cell populations (32, 33), neuronal (34-38), glial (36, 38-41) and/or mesodermal structures (42). Glial tumors [mainly astrocytomas (ASTRs)] and especially glioblastomas are characterized by hypercellularity, pleomorphism, a number of cell mitoses, CIP heterogeneity, various grades of necrosis and multiple endothelial cell proliferations related to the newly generated, tumor-related capillaries (43-65). Furthermore, glial tumors are characterized by high grade local invasiveness and a relatively low metastatic tendency. The specific aims of the present immunohistochemical study were: i) to detect overexpression of COX-2 receptors in primary childhood brain tumors; and ii) to observe the distribution pattern of COX-2 receptors.

Materials and Methods Tissues and tissue handling. In this immunohistochemical study, the following formalin-fixed, paraffin wax-embedded tissue sections of human primary childhood brain tumors were used: i) MEDs/PNETs and ii) different subtypes of ASTRs, all purchased from DAKO Corporation (Carpenteria, CA, USA). The diagnoses of the specific subtypes of the gliomas, observed in this study were established according to WHO guidelines for the classification of glial tumors by a clinical neurohistopathologist (66-70). The technical details of the immunohistochemical techniques employed in this study have

520

already been elaborated by other investigators (71-78) and in the immunohistochemical studies published by our group (79-85). Monoclonal antibody. Mouse anti-COX-2 (Cat. #35-8200; Zymed Laboratories, Inc. South San Francisco, CA, USA), clone; COX 229, Isotype: Mouse IgG1-k was employed. Staining of formalin-fixed and paraffin wax-embedded tissue sections required an antigen retrieval technique, employing preliminary boiling in 10 mM citrate buffer, pH 6.0 (NeoMarkers, Cat. # AP-9003) for 10-20 min, followed by cooling at room temperature for 20 minutes. Immunohistochemical controls. In order to ensure the specificity of the antibody employed in this study, the immunoreactivity of several normal human control tissues including, brain, adrenal, heart, stomach, small intestine, large intestine, liver, kidney, pancreas, lung, testis, ovary, uterus, prostate, thyroid and spleen, all included in one checkerboard multitissue block (DAKO Corporation, Carpinteria, CA, USA; code # T1065) were tested (86, 87). Several postnatal human thymic specimens were also used as negative and positive tissue controls. A number of neoplastically transformed tissues, including malignant melanoma and lung cancer tissues, also represented positive tissue controls. Additional controls for all tissues and MoABs included: i) omission of the primary antibody; ii) utilization of only the enzymatic developer solution to detect the presence of endogenous alkaline phosphatase activity; and iii) utilization of MOPC 21 mouse myeloma IgG1 (ICN) as a replacement for the primary MoAB to determine nonspecific myeloma protein binding to the antigen epitopes of the screened tissues. Immunoalkaline phosphatase antigen detection technique. We used the following immunoalkaline phosphatase cytochemical method (71-78), modified, for antigen detection in formalin-fixed, paraffin-wax embedded anaplastic ASTR and glioblastoma tissues. The technique is a highly sensitive, indirect, four-to-sixstep immunocytochemical method, which combines the biotinstreptavidin-based ABC-method with enzyme-linked (alkaline phosphatase-AP) immunohistochemistry and has been previously described in detail (79-88). Briefly, following deparaffinization in three changes of Xylene substitute (Shandon-Lipshaw, Pittsburgh, PA, USA) for 20 to 30 min, rehydration was carried out employing descending dilutions of alcohol (100% to 50%) to TBS. An initial blocking step using 1% glacial acetic acid mixed with the working buffer for 10 minutes was necessary to eliminate the endogenous AP activity from the tissues. The use of levamisole solution is also described in our earlier observations. As previously described (79-88), GAA inhibition was preferred because of the possible presence of levamisole-resistant AP isoenzyme (73). The second blocking step was conducted with a purified mixture of proteins (Shandon-Lipshaw) from various species for 5-10 min to block cross-reactive antigenic epitopes. Excess serum was removed from the area surrounding the sections. The tissue sections were then incubated for 60-120 min with the COX-2 primary antibody. Next, incubation with the secondary antibody, goat anti-mouse IgG molecule (IgG molecule diluted by ICN Biomedicals, Inc., Aurora, OH, USA), was carried out for 20 min. Streptavidin conjugation was accomplished by incubation with AP conjugated streptavidin for 20 min (75-78). Color visualization of the primary antigen-antibody (Ag-Ab)

Bodey and Kaiser: COX-2 Expression in Childhood Brain Tumors

reaction was accomplished with an alkaline phosphatase (AP) kit I (Vector Laboratories, Burlingame, CA, USA), which contains AS-TR with Tris-HCl buffer at pH 8.2, added for 28-40 min to allow formation of a stable red precipitate. Sections were counterstained with a diluted solution of Gill's hematoxylin (Richard-Allan, Kalamazoo, MI, USA). The tissue slides were then dehydrated in ascending concentrations of alcohol (60% to 100%) to xylene substitute (Shandon-Lipshaw), in which they were kept overnight to ensure complete morphological clearing. The stained tissue sections were mounted using a solution specially designed for use following morphological clearing in xylene substitute (Shandon-Lipshaw). Immunohistochemical evaluation. A qualitative and quantitative evaluation of the percent of antigen-positive cells and the intensity of IGF immunostaining were conducted employing a light microscope (Olympus, Japan) to count 100-200 cells. Artifacts were avoided, while, on the other hand, morphologically characteristic areas were sought out. The presence of brain tumor cells and the endothelial elements of small blood vessels, tumor infiltrating leukocytes and macrophages (the host's immunological effector cells) required careful qualitative assessment. Non-vascular elements were also examined, but only morphologically distinct brain tumor cells were scored. Quantitative evaluation (88): The following evaluations were used: (++++) over 90% of the total brain tumor cells are positive; (+++) 50% to 90% of the total tumor cells are positive; (++) 10% to 50% of the total tumor cells are positive; (+) 1% to 10% of the total brain tumor cells are positive; (±) under 1% of the total tumor cells are positive; (–) negative. Qualitative evaluation (88): The following scores were used: (A) very intense red staining; (B) strong red staining; (C) light red staining; (D) negative staining.

Results In our systematic immunohistochemical screening, the overexpression profile of COX-2 receptors was observed in 14 MEDs/PNETs and 14 subtypes of astrocytic tumors. All 14 MEDs/PNETs observed demonstrated high levels of immunoreactivity (overexpression), detectable by the highest immunostaining intensity grades A and B (Figure 1). In the MEDs/PNETs, 30 to 50% (++) of the tumor cells demonstrated strong presence of the COX-2 receptor. In the observed ASTR subtypes, the COX-2 receptor expression level demonstrated significant differences between the lowgrade pilocytic ASTRs and the high-grade anaplastic ASTRs and glioblastomas. In two low-grade pilocytic ASTRs, the expression level never exceeded 20% and was usually between 5 to 15% (+). On the contrary, in high-grade glial tumors (6 anaplastic ASTRs and 6 GMs) 30 to 50% (++) of the tumor cells overexpressed COX-2 receptors.

Discussion It is well established that dysregulation and altered functioning of various molecular biological processes and signaling pathways are involved in the development and

progression of malignant neoplasms, including brain tumors (89). Employment of chemotherapy in combination with radiotherapy has emerged a strategy of choice in the antineoplastic treatment of advanced human neoplasms (90). COX-2, the isoform of the enzyme cyclooxygenase that is inducible by cytokines, mitogens and growth factors (91), has become one of the principal targets of current antineoplastic chemotherapies. COX-2 is often overexpressed in premalignant and malignant lesions and usually signals a more aggressive pathobiological tumor behavior. The results of the present immunohistochemical study confirm the overexpression of COX-2 in primary human childhood brain tumors, as well as its differential expression in ASTR subtypes. COX-2 has been shown to induce resistance in neoplastically transformed cells to chemotherapeutic agents or radiation. In vitro experimental systems have revealed that selective COX-2 inhibitors cause a decrease in cell proliferation, an increase spontaneous cell death (apoptosis) and modulate cell cycle regulations at gene levels. Molecular targeting the COX-2 enzyme with selective inhibitors was found to be a potent enhancer of neoplastic radioresponse in several preclinical models (92). In fact, COX-2 inhibitors that have been studied in cancer chemoprevention are now under active investigation for cancer therapy (93). In order to maximize the benefit of COX-2 inhibitor therapy in the clinical setting, it is important to investigate the role of molecular targets of such therapy both in established human cancers and in pre-neoplastic lesions, as well as in its effects on currently used therapeutic modalities such as radiotherapy (94). Aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) are the most widely studied agents for the chemoprevention of several human malignancies (95, 96). These compounds exert their effects by a number of mechanisms. They inhibit both COX-1 and COX-2 (97). When treated with sulindac, an NSAID that inhibits both COX-1 and COX-2, the number of intestinal adenomas is reduced by more than 90% (98) and the total volume of colon tumors by more than 52% (99). This reduction in polyp formation can be improved by use of a more selective COX-2 inhibitor or by the deletion of the COX-2 gene itself (100, 101). The mechanisms by which the inhibition of COX-2 leads to decreased colon carcinogenesis are not fully understood, but may involve an increase in apoptosis (101, 102), the regulation of angiogenesis, or both (103, 104). Aspirin and other NSAIDs may also act by COX-independent mechanisms, such as inhibition of the activation of nuclear factor-κB (NF-κB) (105) or interference with the binding of the peroxisome-proliferator–activated receptor (PPAR ) to DNA (106). A selective COX-2 inhibitor, celecoxib, was shown in a randomized study to cause regression of polyps in patients with familial adenomatous polyposis. Celecoxib significantly reduced the number of colorectal polyps by 28% after 6 months of treatment, compared with patients administered

521

in vivo 20: 519-526 (2006)

Figure 1. Childhood medulloblastoma. A great majority of the neoplastically-transformed cells (+++) demonstrated strong expression (B) of COX-2 receptor, with focal overexpression documented by very strong staining (A). Routine, 10% buffered formalin fixation. Paraffin-wax embedding. Alkaline phosphatase conjugated streptavidin-biotin antigen detection technique. Magnification: 200x.

placebo (107). Celecoxib currently is being investigated in trials for Barrett’s esophagus and cancers of the bladder, colon, mouth, prostate and skin (108). Sulindac and its derivatives have demonstrated antineoplastic effects–through COX-1, COX-2 and LOX inhibition (109). The LOX pathway in AA metabolism is a promising target for chemoprevention research; LOX inhibition has been shown to reduce tumor cell growth more effectively than COX inhibition (110). LOXs have been associated with various neoplastically transformed cell types, including breast, colon, prostate, pancreas and non-small-cell lung cancers (111, 112). In conclusion, inhibitors of the cyclooxygenases may be yet another component to be considered when developing the "cocktail" that will lead to the most efficacious treatment of neoplastic disease. Based upon our results, the COX-2 receptor may now be added to the long list of brain tumor progression markers and possible therapeutic targets.

522

References 1 2

3

4

5

Taketo MM: Cyclooxygenase-2 inhibitors in tumorigenesis. J Natl Cancer Inst 90: 1529-1536, 1998. Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, Wakabayashi N, Saunders B, Shen Y, Fujimura T, Su LK and Levin B: The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Eng J Med 342: 1946-1952, 2000. Herschman HR, Talley JJ and DuBois R: Cyclooxygenase 2 (COX-2) as a target for therapy and noninvasive imaging. Mol Imaging Biol 5: 286-303, 2003. Lin DW and Nelson PS: The role of cyclooxygenase-2 inhibition for the prevention and treatment of prostate carcinoma. Clin Prostate Cancer 2: 119-126, 2003. Tuynman JB, Hulscher JB, Steller EP, van Lanschot JJ and Richel DJ: Cyclooxygenase(COX)-2-inhibition in the prevention and treatment of colorectal carcinoma. Ned Tijdsch Geneeskd 147: 2207-2212, 2003.

Bodey and Kaiser: COX-2 Expression in Childhood Brain Tumors

6 7 8 9

10

11

12

13

14

15 16

17

18

19 20

21

22

23

Cohen MC and Cohen S: Cytokine function. A study in biologic diversity. Am J Clin Pathol 105: 589-598, 1996. Lunney JK: Cytokines orchestrating the immune response. ReSciTechn Off Int Epiz 17: 84-94, 1998. Fosslien E: Molecular pathology of cyclooxygenase-2 in neoplasia. Ann Clin Lab Sci 30: 3-21, 2000. Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S and DuBois RN: Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology 107: 1183-1188, 1994. Fujita T, Matsui M, Takaku K, Uetake H, Ichikawa W, Taketo MM and Sugihara K: Size- and invasion-dependent increase in cyclooxygenase 2 levels in human colorectal carcinomas. Cancer Res 58: 4823-4826, 1998. Milas L: Cyclooxygenase-2 (COX-2) enzyme inhibitors as potential enhancers of tumor radioresponse. Semin Radiat Oncol 11: 290-299, 2001. Joki T, Heese O, Nikas DC, Bello L, Zhang J, Kraeft SK, Seyfried NT, Abe T, Chen LB, Carroll RS and Black PM: Expression of cyclooxygenase 2 (COX-2) in human glioma and in vitro inhibition by a specific COX-2 inhibitor, NS-398. Cancer Res 60: 4926-4931, 2000. Kirschenbaum A, Liu X, Yao S and Levine AC: The role of cyclooxygenase-2 in prostate cancer. Urology 58: 127-131, 2001. Yang VW, Shields JM, Hamilton SR, Spannhake EW, Hubbard WC, Hylind LM, Robinson CR and Giardiello FM: Size-dependent increase in prostanoid levels in adenomas of patients with familial adenomatous polyposis. Cancer Res 58: 1750-1753, 1998. Allen JC and Siffert J: Contemporary issues in the management of childhood brain tumors. Curr Opin Neurol 10: 137-141, 1997. Zeltzer PM: Chemotherapy for medulloblastoma: A review of treatment in recurrent and de novo tumors. In: Medulloblastomas in Children: New Concepts in Tumor Biology, Diagnosis and Treatment. Zeltzer PM, Pochedly C (eds.). New York, Praeger, pp. 182-199, 1986. Tomita T, McLone DG and Yasue M: Cerebral primitive neuroecto-dermal tumors in childhood. J Neuro-Oncol 6: 233243, 1988. Ilveskoski I, Pihko H, Sankila R, Lanning M, Perkkio M, Aarimaa T, Makipernaa A and Saarinen UM: Improving outcome of malignant brain tumours in very young children: a population-based study in Finland during 1975-93. Acta Paediatrica 86: 724-729, 1997. Kun LE: Brain tumors. Challenges and directions. Pediatric Clin North Am 44: 907-917, 1997. Kuttesch JF Jr: Advances and controversies in the management of childhood brain tumors. Curr Opin Oncol 9: 235-240, 1997. Hartsell WF, Gajjar A, Heideman RL, Langston JA, Sanford RA, Walter A, Jones D, Chen G and Kun LE: Patterns of failure in children with medulloblastoma: effects of preirradiation chemotherapy. Int J Radiat Oncol Biol Phys 39: 15-24, 1997. Bailey P and Cushing H: Medulloblastoma cerebelli, a common type of midcerebellar glioma of childhood. Arch Neurol Psychiatry 14: 192-224, 1925. Schaper A: Einige kritische Bemerkungen zu Lugaro's Aufsatz: "Ueber die Histogenese der Korner der Kleinhirnrinde". Anat Anz 10: 422-426, 1895.

24 Camins MB, Cravieta HM, Epstein F and Ransohoff J: Medulloblastoma: an ultrastructural study – evidence for astrocytic and neuronal differentiation. Neurosurgery 6: 398411, 1980. 25 Packer RJ, Sutton LN, Rorke LB, Littman PA, Sposto R, Rosenstock JG, Bruce DA and Schut L: Prognostic importance of cellular differentiation in medulloblastoma of childhood. J Neurosurg 61: 296-301, 1984. 26 Caputy AJ, McCullough DC, Manz HJ, Patterson K and Hammock MK: A review of the factors influencing the prognosis of medulloblas-toma. The importance of cell differentiation. J Neurosurg 66: 80-87, 1987. 27 Hart MN and Earle KM: Primitive neuroectodermal tumors of the brain in children. Cancer 32: 890-897, 1973. 28 Rorke L: The cerebellar medulloblastoma and its relationship to primitive neuroectodermal tumors. J Neuropathol Exp Neurol 42: 1-15, 1983. 29 Rorke LB: Origin and histogenesis of medulloblastoma. In: Medulloblastomas in Children: New Concepts in Tumor Biology, Diagnosis and Treatment. Zeltzer PM, Pochedly C, (eds.). New York, Praeger, pp. 14-21, 1986. 30 Rorke LB, Gilles FR, Davis RL and Becker LE: Revision of the World Health Organization classification of brain tumors for childhood brain tumors. Cancer 56: 1869-1886, 1985. 31 Rubinstein LJ: A commentary on the proposed revision of the World Health Organization classification of brain tumors for childhood brain tumors. Cancer 56: 1887-1888, 1985. 32 Bodian M and Lawson D: The intracranial neoplastic diseases of childhood: a description of their natural history based on a clinico-pathological study of 129 cases. Br J Surg 40: 368-392, 1953. 33 Matakas F, Cervos-Navarro J and Gullotia F: The ultrastructure of medulloblastomas. Acta Neuropathol 16: 271-284, 1970. 34 Rubinstein LJ: Embryonal central neuroepithelial tumors and their differentiating potential: a cytogenctic view of a complex neuro-oncological problem. J Neurosurg 62: 795-805, 1985. 35 Rubinstein LJ, Herman MM and van der Berg SR: Differentiation and anaplasia in central neuroepithelial tumors. Prog Exp Tumor Res 27: 32-48, 1984. 36 Rubinstein LJ: The cerebellar medulloblastoma: its origin, differentiation, morphological variants, and biological behavior. In: Tumors of The Brain and Skull. Part III. Vincken PJ, Bruyn GW (eds.). New York, Elsevier, pp. 167-194, 1975. 37 Trojanowski JQ, Lee VM-Y and Schlaepfer WW: An immunohis-tochemical study of human central and peripheral nervous system tumors, using monoclonal antibodies against neurofilaments and glial filaments. Hum Pathol 15: 248-257, 1984. 38 Roessmann U, Velasco ME, Gambetti P and Autilio-Gambetti L: Neuronal and astrocytic differentiation in human neuroepithelial neoplasms. J Neuropath Exp Neurol 42: 113-121, 1983. 39 Gullotta F and Kersting G: The ultrastructure of medulloblastoma in tissue culture. Virchows Arch (Pathol Anat) 356: 111-118, 1972. 40 Palmer JO, Kasselberg AG and Netsky MG: Differentiation of medulloblastoma: studies including immunohistochemical localization of glial fibrillary acidic protein. J Neurosurg 55: 161169, 1981. 41 Roy S: An ultrastructural study of medulloblastoma. Neurol India 25: 226-229, 1977.

523

in vivo 20: 519-526 (2006) 42 Dickson DW, Hart MN, Menezes A and Cancilla PA: Medulloblastoma with glial and rhabdomyoblastic differentiation. J Neuropathol Exp Neurol 42: 639-647, 1983. 43 Kemohan JW, Mabon RF, Svien YU and Adson AW: A simplified classification of gliomas. Proc Staff Meet Mayo Clin 24: 71-75, 1949. 44 Ringertz J: Grading of gliomas. Acta Pathol Microbiol Scand 27: 51-64, 1950. 45 Kemohan JW and Sayre GP: Tumors of Central Nervous System. Fascicle 35, Atlas of Tumor Pathology. Armed Forces Institute of Pathology, Washington, pp. 17-129, 1952. 46 Zülch KJ: Histological typing of tumors of central nervous system. International histological classification of tumours, World Health Organisation, Geneva 21: 1-56, 1979. 47 Nelson JS, Tuskada Y, Schoenfeld D, Fulling K, Lamarche J and Peress N: Necrosis as a prognostic criteria in malignant supratentorial astrocytic gliomas. Cancer 52: 550-554, 1983. 48 Burger PC, Vogel FS, Green SB and Strike AT: Glioblastoma multiforme and anaplastic astrocytoma. Pathologic criteria and prognostic implications. Cancer 56: 1106-1111, 1985. 49 DeArmond SJ: In situ cell kinetic studies on human neuroectodermal tumors with bromodeoxyuridine labeling. J Neurosurg 64: 453-459, 1986. 50 Shapiro JR: Biology of gliomas. Heterogenity, oncogenes, growth factors. Semin Oncol 133: 4-15, 1986. 51 Hoshino T, Rodriguez LA, Cho KC, Lee KS, Wilson CB, Edwards MSB, Levin VA and Davis RL: Prognostic implications of the proliferative potential of low grade astrocytomas. J Neurosurg 69: 839-842, 1988. 52 Hoshino T, Prados M, Wilson CB, Cho KG, Lee KS and Davis RL: Prognostic implications of low bromodeoxyuridine labeling index of human gliomas. J Neurosurg 71: 335-341, 1989. 53 Burger PC and Kleihues P: Cytologic composition of the untreated glioblastoma with implications for evaluation of needle biopsies. Cancer 63: 2014-2023, 1989. 54 Russel DS and Rubinstein LJ: Tumors of neuroepithelial origin. In: Pathology of Tumours of the Nervous System. 5th Edition, Edward Arnold, London, pp. 83-350, 1989. 55 Bodey B, Zeltzer PM, Saldivar V and Kemshead J: Immunophenotyping of childhood astrocytomas with a library of monoclonal antibodies. Int J Cancer 45: 1079-1087, 1990. 56 Weller RO: Tumours of the nervous system. In: Systemic Pathology, 3rd Edition. Volume IV. Symmers SC (ed). Churchill Livingston, Edinburgh, pp. 427-503, 1990. 57 Schmitt BP, Oberwittler C and Schad LR: Computer aided classification of malignancy in astrocytomas: I. The value of nuclear parameters obtained by automated black and white image analysis. Analyt Cell Pathol 4: 397-407, 1992. 58 Schmitt HP and Oberwittler C: Computer aided classification of malignancy in astrocytomas II. The value of categorically evaluating histologic and nonhistologic features for a numerical classifier. Analyt Cell Pathol 4: 409-419, 1992. 59 Kleihues P, Burger PC and Scheithauer BW: Histological Typing of Tumors of the Central Nervous System. International Histological Classification of Tumours. 2nd Edition, World Health Organisation, Springer Verlag, Berlin, 1993. 60 Dinda AK, Kharbanda K, Sarkar C, Roy S, Mathur M and Banedi AK: In vivo proliferative potential of primary human brain tumors. Its correlation with histological classification and morphological. I. Gliomas. Pathology 25: 4-9, 1993.

524

61 Dinda AK, Kharbanda K, Sarkar C, Roy S, Mathur M and Banedi AK: In vivo proliferative potential of primary human brain tumors. Its correlation with histological classification and morphological features. II. Nonglial Tumors. Pathology 25: 1014, 1993. 62 Burger PC and Scheithauer BW: Tumors of the Central Nervous System. Armed Forces Institute of Pathology, Washington, 1994. 63 Sharma S, Karak AK, Sarkar C, Gomathy G, Bannedi AK and Schmitt HP: A grading study of gliomas using histologic morphometry and malignancy classification. J Neurol Oncol 27: 75-85, 1996. 64 Bodey B, Gröger AM, Bodey B Jr, Siegel SE and Kaiser HE: Immunocytochemical detection of p53 protein expression in various childhood astrocytoma subtypes: significance in tumor progression. Anticancer Res 17: 1187-1194, 1997. 65 Louis DN: A molecular genetic model of astrocytoma histopathology. Brain Pathol 7: 755-764, 1997. 66 Zülch KJ and Wechsler W: Pathology and Classification of Gliomas. Progr Neurol Surg, Vol. 2, Karger, Basel-New York, pp. 1-84, 1968. 67 Zülch KJ: Histologic Classification of Tumours of the Central Nervous System. International Histological Classification of Tumours, No. 21, World Health Organization, Geneva, 1979. 68 Zülch KJ: Principles of the new World Health Organization (WHO) classification of brain tumors. Neuroradiology 19: 5966, 1980. 69 Szymas J: Histologic classification of central nervous system tumors by the World Health Organization. Pol J Pathol 45: 8191, 1994. 70 Szymas J: Diagnostic immunohistochemistry of tumors of the central nervous system. Folia Neuropathol 32: 209-214, 1994. 71 Janckila AJ, Yam LT and Li C-Y: Immunoalkaline phosphatase cytochemistry. Am J Clin Pathol 84: 476-480, 1985. 72 Cattoretti G, Berti E, Schiro R, D' Amato L, Valeggio C and Rilke F: Improved avidin-biotin-peroxidase complex (ABC) staining. Histol J 20: 75-80, 1988. 73 Yam LT, Janckila AJ, Epremian BE and Li C-Y: Diagnostic significance of levamisole-resistant alkaline phosphatase in cytochemistry and immunocytochemistry. Am J Clin Pathol 91: 31-36, 1989. 74 Strasburger CJ, Amir-Zaltsman Y and Kohen F: The avidinbiotin reaction as an universal amplification system in immunoassays. Prog Clin Biol Res 285: 79-100, 1988. 75 Wilchek M and Bayer EA: Introduction to avidin-biotin technology. Methods Enzymol 184: 5-13, 1990. 76 Duhamel RC and Whitehead JS: Prevention of nonspecific binding of avidin. Methods Enzymol 184: 201-207, 1990. 77 Diamandis EP and Christopoulos TK: The biotin(strept)avidin system: principles and applications in biotechnology. Clin Chem 37: 625-636, 1991. 78 Hsu SM, Raine L and Fanger H: Use of avidin-biotinperoxidase complex (ABC) in immunoperoxidase techniques: a comparison of ABC and unlabeled antibody (PAP) procedure. J Histochem Cytochem 29: 577-581, 1981. 79 Bodey B, Bodey B Jr, Siegel SE and Kaiser HE: Immunocytochemical detection of the Homeobox B3, B4 and C6 gene products in childhood medulloblastomas/primitive neuroectodermal tumors. Anticancer Res 20: 1769-1780, 2000.

Bodey and Kaiser: COX-2 Expression in Childhood Brain Tumors

80 Bodey B, Bodey B Jr, Siegel SE and Kaiser HE: Fas (APO-1, CD95) receptor expression in childhood astrocytomas. Is it a marker of the major apoptotic pathway or a signaling receptor for immune escape of neoplastic cells? In Vivo 13: 357-373, 1999. 81 Bodey B, Bodey B Jr, Siegel SE and Kaiser HE: Matrix metalloproteinase expression in childhood medulloblastomas/ primitive neuroectodermal tumors. In Vivo 14: 667-673, 2000. 82 Bodey B, Bodey B Jr, Siegel SE and Kaiser HE: Significant differences in the matrix metalloproteinase expression profiles of spontaneous medulloblastomas/primitive neuroectodermal tumors as compared with their xenografted, established tumor cell line-derived counterparts. In Vivo 14: 675-682, 2000. 83 Bodey B, Bodey B Jr, Siegel SE and Kaiser HE: Immunocytochemical detection of leukocyte-associated and apoptosis-related antigen expression in childhood brain tumors. Crit Rev Oncol/Hematol 39: 3-16, 2001. 84 Bodey B: The significance of immunocytochemistry in the diagnosis and therapy of neoplasms. Expert Opin Biol Ther 2: 371-393, 2002. 85 Bodey B, Siegel SE and Kaiser HE: MAGE-1, a cancer-testis antigen, expression in childhood astrocytomas as an indicator of tumor progression. In Vivo 16: 583-588, 2002. 86 Battifora H: The multitumor (sausage) tissue block: novel method for immunohistochemical antibody testing. Lab Invest 55: 244-248, 1986. 87 Battifora H and Mehta P: The checkerboard tissue block. An improved multitissue control block. Lab Invest 63: 722-724, 1990. 88 Bodey B, Zeltzer PM, Saldivar V and Kemshead J: Immunophenotyping of childhood astrocytomas with a library of monoclonal antibodies. Int J Cancer 45: 1079-1087, 1990. 89 Liao Z and Milas L: Cox-2 and its inhibition as a molecular target in the prevention and treatment of lung cancer. Expert Rev Anticancer Ther 4: 543-560, 2004. 90 Nakata E, Mason KA, Hunter N, Husain A, Raju U, Liao Z, Ang KK and Milas L: Potentiation of tumor response to radiation or chemoradiation by selective cyclooxygenase-2 enzyme inhibitors. Int J Radiat Oncol Biol Phys 58: 369-375, 2004. 91 Cao Y and Prescott SM: Many actions of cyclooxygenase-2 in cellular dynamics and in cancer. J Cell Physiol 190: 279-286, 2002. 92 Milas L, Mason KA, Liao Z and Ang KK: Chemoradiotherapy: emerging treatment improvement strategies. Head Neck 25: 152167, 2003. 93 Sabichi AL and Lippman SM: COX-2 inhibitors and other NSAIDs in bladder and prostate cancer. Prog Exp Tumor Res 37: 163-178, 2003. 94 Choy A and Milas L: Enhancing radiotherapy with cyclooxygenase enzyme inhibitors: a rational advance? J Natl Cancer Inst 95: 1440-1452, 2003. 95 Shiff SJ and Rigas B: Nonsteroidal anti-inflammatory drugs and colorectal cancer: evolving concepts of their chemopreventive actions. Gastroenterology 113: 1992-1998, 1997. 96 Kimmey MB and Lanas A: Review article: Appropriate use of proton pump inhibitors with traditional nonsteroidal antiinflammatory drugs and COX-2 selective inhibitors. Aliment Pharmacol Ther 19: 60-65, 2004. 97 Taketo MM: Cyclooxygenase-2 inhibitors in tumorigenesis. J Natl Cancer Inst 90: 1529-1536, 1998.

98 Boolbol SK, Dannenberg AJ, Chadburn A, Martucci C, Guo XJ, Ramonetti JT, Abreu-Goris M, Newmark HL, Lipkin ML, DeCosse JJ and Bertagnolli MM: Cyclooxygenase-2 overexpression and tumor formation are blocked by sulindac in a murine model of familial adenomatous polyposis. Cancer Res 56: 2556-2560, 1996. 99 Rao CV, Rivenson A, Simi B, Zang E, Kelloff G, Steele V and Reddy BS: Chemoprevention of colon carcinogenesis by sulindac, a nonsteroidal anti-inflammatory agent. Cancer Res 55: 1464-1472, 1995. 100 Kawamori T, Rao CV, Seibert K and Reddy BS: Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis. Cancer Res 58: 409-412, 1998. 101 Sheng H, Shao J, Morrow JD, Beauchamp RD and DuBois RN: Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res 58: 362-366, 1998. 102 Chan TA, Morin PJ, Vogelstein B and Kinzler KW: Mechanisms underlying nonsteroidal antiinflammatory drug-mediated apoptosis. Proc Natl Acad Sci USA 95: 681-686, 1998. 103 Jones MK, Wang H, Peskar BM, Levin E, Itani RM, Sarfeh IJ and Tarnawski AS: Inhibition of angiogenesis by nonsteroidal anti-inflammatory drugs: Insight into mechanisms and implications for cancer growth and ulcer healing. Nat Med 5: 1418-1423, 1999. 104 Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M and DuBois RN: Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 93: 705-716, 1998. 105 Yamamoto Y, Yin MJ, Lin KM and Gaynor RB: Sulindac inhibits activation of the NF-kappaB pathway. J Biol Chem 274: 27307-27314, 1999. 106 He T-C, Chan TA and Vogelstein B: PPARdelta is an APCregulated target of nonsteroidal anti-inflammatory drugs. Cell 99: 335-345, 1999. 107 Steinbach G, Lynch PM and Phillips RKS: The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Eng J Med 342: 1946-1952, 2000. 108 Kelloff GJ, Sigman CC and Greenwald P: Cancer chemoprevention: progress and promise. Eur J Cancer 35: 17551762, 1999. 109 Haanen C: Sulindac and its derivatives: a novel class of anticancer agents. Curr Opin Investig Drugs 2: 677-683, 2001. 110 Hong SH, Avis I, Vos MD, Martinez A, Treston AM and Mulshine JL: Relationship of arachidonic acid metabolizing enzyme expression in epithelial cancer cell lines to the growth effect of selective biochemical inhibitors. Cancer Res 59: 22232228, 1999. 111 Gupta S, Srivastava M, Ahmad N, Sakamoto K, Bostwick DG and Mukhtar H: Lipoxygenase-5 is overexpressed in prostate adenocarcinoma. Cancer 91: 737-743, 2001. 112 Moody TW, Leyton J, Martinez A, Hong S, Malkinson A and Mulshine JL: Lipoxygenase inhibitors prevent lung carcinogenesis and inhibit non-small cell lung cancer growth. Exp Lung Res 24: 617-628, 1998.

Received December 28, 2005 Accepted April 27, 2006

525