child cns

Upload: krishanbansal2247

Post on 30-May-2018

212 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/9/2019 Child CNS

    1/16

    INVITED PAPER

    Current concepts in the molecular genetics of pediatric

    brain tumors: implications for emerging therapies

    Mandeep S. Tamber & Krishan Bansal & Muh-Lii Liang &

    Todd G. Mainprize & Bodour Salhia & Paul Northcott &

    Michael Taylor & James T. Rutka

    Received: 14 March 2006 / Published online: 2 September 2006# Springer-Verlag 2006

    AbstractBackground The revolution in molecular biology that has

    taken place over the past 2 decades has provided research-

    ers with new and powerful tools for detailed study of the

    molecular mechanisms giving rise to the spectrum of

    pediatric brain tumors. Application of these tools has

    greatly advanced our understanding of the molecular

    pathogenesis of these lesions.

    Review After familiarizing readers with some promising

    new techniques in the field of oncogenomics, this review

    will present the current state of knowledge as it pertains to

    the molecular biology of pediatric brain neoplasms. Along

    the way, we hope to highlight specific instances where thedetailed mechanistic knowledge acquired thus far may be

    exploited for therapeutic advantage.

    Keywords Brain neoplasms . Molecular biology .

    Pediatrics . Review . Therapeutics

    Introduction

    In the past 2 decades, the textbooks on the molecular

    biology of pediatric brain tumors have been rewritten

    several times. This has been principally due to the

    tremendous advances that have been made in our under-

    standing of these tumors from a basic science standpoint,

    a dv an ce s which a re in large p art d ue to the n ew

    technologies that have come forward to query the genetic

    underpinnings of these lesions. Suffice it to say that we are

    now poised, greater than ever before, to utilize the

    information that has been garnered toward tangible

    improvements in the way children with brain tumors are

    treated.

    In this review, we will summarize the salient molecular

    genetic changes that characterize pediatric astrocytomas,

    both low- and high-grade lesions, ependymoma, medullo-

    blastoma and primitive neuroectodermal tumors, atypical

    teratoid rhabdoid tumor (AT/RT), germ cell tumors (GCTs),

    and sarcomas. During the review of each tumor type, we

    will attempt to provide clues as to how detailed knowledge

    of the molecular pathogenesis of these lesions may be

    harnessed toward translational research opportunities

    designed to improve patient outcomes.

    New techniques in the field of oncogenomics

    In the past 30 years, advanced cancer cytogenetics have

    been applied to numerous cancer subtypes, including many

    of the pediatric brain tumors. For additional details

    regarding these techniques, the interested reader is referred

    to several excellent review articles on this topic [ 136, 159].

    Here we will describe array comparative genomic hybrid-

    Childs Nerv Syst (2006) 22:13791394

    DOI 10.1007/s00381-006-0187-3

    M. S. Tamber: K. Bansal : M.-L. Liang : T. G. Mainprize :

    B. Salhia: P. Northcott: M. Taylor: J. T. Rutka

    Division of Neurosurgery, The Hospital for Sick Children,

    The University of Toronto,

    Toronto, Ontario, Canada

    M. S. Tamber: K. Bansal : M.-L. Liang : T. G. Mainprize :

    B. Salhia: P. Northcott: M. Taylor: J. T. Rutka

    Arthur and Sonia Labatt Brain Tumor Research Centre,

    The Hospital for Sick Children, The University of Toronto,

    Toronto, Ontario, Canada

    J. T. Rutka (*)

    The Division of Neurosurgery, Suite 1504,

    The Hospital for Sick Children,

    555 University Avenue,

    Toronto, Ontario 518, Canada

    e-mail: [email protected]

  • 8/9/2019 Child CNS

    2/16

    ization (CGH), single nucleotide polymorphism (SNP)

    arrays, microarray analysis of gene expression, and exon

    resequencing as new techniques which have been and will

    be applied to the study of pediatric brain tumors (Table 1).

    Array comparative genomic hybridization

    Array comparative genomic hybridization is similar toCGH, but instead of hybridizing the labeled probes

    to metaphase chromosomes, the probes are hybridized to

    microarray slides in which thousands of fragments of DNA

    that span the genome have been arrayed out [29, 70, 114,

    115]. The array CGH chip can be spotted with DNA

    composed of genomic clones, cDNA sets, or oligonucleo-

    tide probes depending on the array design. A major

    advantage of array CGH over traditional CGH is the

    significantly higher resolution offered by the array plat-

    form, allowing investigators to identify smaller areas of

    aberration. For example, bacterial artificial chromosome

    (BAC) arrays consisting of greater than 30,000 BAC clonesrepresenting the entire human genome are now widely used

    in array CGH, permitting whole-genome copy number

    analysis at resolutions of less than 1 Mb, compared with the

    much lower resolution achieved using traditional CGH

    (~5 Mb) [72, 83].

    The highest resolution for array CGH is now provided

    by genome tiling arrays which interrogate entire genomes

    with evenly spaced oligonucleotide probes [8, 19]. The

    increased resolution offered by modern array CGH plat-

    forms will allow for the incredibly fine mapping of focal

    amplifications and deletions not possible using previous

    generation arrays. Weaknesses of array CGH include an

    inability to detect structural anomalies such as reciprocaltranslocations and the inability to detect copy-number-

    neutral loss of heterozygosity (LOH) events.

    Array CGH technology has recently been applied by a

    number of groups studying DNA copy number changes in

    pediatric brain tumors [24, 68, 99, 135, 161, 164]. Using a

    combination of array CGH and oligonucleotide gene

    expression arrays (discussed below) in a study of ependy-

    moma, Taylor et al. recently reported that histologically

    similar anatomical subtypes of ependymoma could be

    distinguished based upon their distinct patterns of copy-

    number-driven gene expression [161]. These authors also

    used array CGH and expression microarrays to identify anumber of genes that are both amplified and overexpressed

    in ependymoma (i.e., oncogenes). The results of this study

    emphasize the power of array CGH in the identification of

    genomic aberrations in cancer and demonstrate its utility

    when combined with complementary technical platforms

    such as gene expression arrays.

    Table 1 Summary of molecular and cytogenetic techniques discussed in the text

    Technique CGH SKY Array CGH SNP array Expression

    array

    Exon

    resequencing

    Primary

    application

    Mapping regions

    of amplification

    and deletion

    Identification of

    structural

    aberrations

    Copy number

    analysis

    Copy number

    analysis; LOH

    mapping

    Gene

    expression

    profiling

    Mutation

    detection

    Platform type Fluorescence

    microscopy

    Fluorescence

    microscopy

    Microarray:

    BAC,

    oligonucleotide

    Microarray:

    oligonucleotide

    Microarray:

    oligonucleotide

    Dideoxy

    sequencing

    Starting

    material

    Genomic DNA Metaphase

    chromosomes from

    cultured cells

    Genomic DNA Genomic DNA Total RNA or

    mRNA

    Genomic DNA

    Advantages Assay only

    requires genomic

    DNA

    Capacity to detect

    balanced

    translocations

    Good resolution;

    whole-genome

    screening

    High

    resolution;

    whole-genome

    screening

    Whole

    transcriptome

    screening

    possible

    Highest

    resolution;

    no restriction

    on choice of

    genes

    Disadvantages Poor resolution;

    unable to detect

    balanced

    translocations

    Prone to erroneous

    interpretations and

    classification errors

    LOH mapping

    not possible

    Data analysis

    intensive

    Data analysis

    and

    interpretation

    issues

    Limited to user-

    defined genes

    and gene

    families;

    expensive

    Listed are some of the current platforms employed in cancer genetics to identify novel disease-causing genes, highlighting the type of platform

    used, biological material required, as well as primary strengths and weaknesses of each technique. Detailed information for each assay type,

    including relevant clinical examples, is described in the text.

    CGH comparative genomic hybridization, SKY spectral karyotyping, SNP single nucleotide polymorphism, LOH loss of heterozygosity,

    BAC bacterial artificial chromosome

    1380 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    3/16

    Single nucleotide polymorphism arrays

    Single nucleotide polymorphisms represent the most com-

    mon form of genetic variation in the human genome [2, 62,

    137, 169]. SNPs are naturally occurring variations in DNA

    sequence that occur regularly throughout the genome, with

    an average frequency of one SNP for every 200 bases of

    DNA. Recently, array-based technologies that use SNPs asa means of investigating both DNA copy number and SNP

    genotype have become commercially available [67, 79, 95,

    96]. Unlike array CGH which involves simultaneous

    cohybridization of both a test sample and a reference

    sample to the microarray, SNP arrays hybridize only a test

    sample, with DNA copy number calculated by comparison

    of signal intensity data obtained from a test sample to data

    from control samples hybridized to the same array type. In

    SNP array analysis, DNA from a test sample is first

    digested with a specific restriction enzyme (e.g., Hind or

    Xba) and then ligated to an adaptor which permits

    amplification of the digested fragments. Amplified DNA

    is subsequently fragmented and labeled before hybridiza-

    tion to a specific SNP array (e.g. 50K Hind or 50K Xba

    array) (Fig. 1).The arrays consist of 25-mer oligonucleotide probes

    that are designed to genotype the test sample for either of

    two alleles of a given SNP (arbitrarily designated A and B

    alleles). Each SNP is represented by a set of perfect match

    A and perfect match B probes, as well as an equal number

    of mismatch probes for both alleles. For any given SNP,

    the possible genotypes for a test sample include AA, BB,

    Fig. 1 Analysis of LOH and chromosomal copy number using SNP

    arrays. Data obtained from SNP array experiments can be analyzed

    visually to identify instances of LOH and copy number gains and

    losses. a Inferred LOH view of a medulloblastoma cell line exhibiting

    genetic loss on chromosome 17p. The middle panel depicts inferred

    regions of LOH in blue and those retaining heterozygosity in yellow.

    The significance curve shown on the right demonstrates a high

    probability of LOH on 17p for this sample. b Copy number view of a

    medulloblastoma cell line with high-level genomic amplifications on

    chromosome 8q24. Copy number is displayed in the middle panel

    using a user-defined scale, with gains and losses represented by darker

    and lighter color intensities, respectively. The right panel clearly

    identifies the 8q24 amplifications in this sample as sharp peaks in copy

    number, with values dramatically exceeding the threshold value of 2 at

    these loci

    Childs Nerv Syst (2006) 22:13791394 1381

  • 8/9/2019 Child CNS

    4/16

    or AB, with genotype determined by the pattern of perfect

    match and mismatch signal intensities obtained for that

    SNP. Accuracy of SNP genotyping relies on the availabil-

    ity of a highly purified DNA sample; analysis of tumor

    tissue may therefore be problematic due to the presence of

    contaminating normal cells. Modern array platforms

    interrogate from 100,000 to 500,000 SNPs, resulting in

    high-resolution genome coverage with median SNPspacing of ~8.5 and ~2.5 kb, respectively. This resolution

    is more than 1,000 times better than classic CGH. As a

    result of the high resolution and sensitivity associated

    with SNP arrays, it is possible to identify microamplifi-

    cations and microdeletions present in cancer samples, as

    well as larger aberrations including aneuploidy. A major

    advantage of SNP array technology over other platforms

    used in cancer genetics is the simultaneous acquisition of

    both copy number and genotype information. Whereas

    copy number data are critical for mapping regions of

    amplification and homozygous deletion in tumor samples,

    genotyping information can identify loci which haveundergone loss of heterozygosity without a decrease in

    copy number (copy-number-neutral LOH). Although

    SNP-array-based approaches have not yet been reported

    for pediatric brain tumors, they have proven to be a

    valuable tool when applied to studies of other tumor types

    [49, 65, 140, 162, 176, 179, 180].

    Microarray analysis of gene expression

    In recent years, microarrays designed to study global gene

    expression profiles have become an indispensable tool for

    cancer researchers [16, 127, 142]. Significant improve-

    ments with respect to array design, in combination with a

    more comprehensive knowledge of the human transcrip-

    tome, has resulted in the evolution of arrays which allow

    for evaluation of the expression of every known human

    gene [47, 60, 87, 88, 141]. In addition, new-generation

    high-resolution arrays have recently been developed,

    including platforms which interrogate every annotated

    human exon. Now, genome tiling arrays capable of

    measuring gene expression every 25 base pairs across

    entire genome have been developed in order to measure

    expression from cryptic transcriptional units [21, 75, 78].

    This technology has found expression of countless tran-

    scripts that previously lacked annotation, many from so-

    called gene deserts.

    Conventional arrays for gene expression analysis consist

    of 25-mer oligonucleotide probes that are complementary in

    sequence to their target, with multiple perfect match and

    mismatch probes existing for each interrogated target and

    multiple probe sets present for each gene represented on the

    array. Total RNA from a tumor sample is first reverse-

    transcribed using a T7-oligo(dT) promoter primer to

    generate first-strand cDNA. First-strand cDNA is then

    converted to double-stranded cDNA which serves as a

    template for the synthesis of complementary RNA (cRNA)

    in an in vitro transcription (IVT) reaction. IVT is carried out

    by T7 RNA polymerase in the presence of biotin-labeled

    ribonucleotides. The biotinylated cRNA is subsequently

    purified, fragmented, and hybridized to an expression array.

    Hybridized arrays are then stained, washed, and scanned todetermine the signal intensity for each probe on the array,

    with the signal intensity being proportional to the amount

    of bound target. Data obtained from such a gene expression

    array can then be used to identify the gene expression

    profile characteristic of a particular tumor [100]. Gene

    expression arrays have proven useful in the identification of

    aberrantly expressed genes and gene families in pediatric

    brain tumors, with some of these candidates potentially

    representing important prognostic markers and/or serving

    as targets for future therapeutics [44, 51, 92, 119].

    Exon resequencing

    Advances in DNA sequencing technologies, along with the

    availability of comprehensive genomic databases, have

    recently led to a number of large-scale resequencing efforts

    aimed at identifying novel mutations in groups or families

    of preselected candidate genes [7, 27, 28, 122, 145, 146,

    170, 171]. The resequencing approach involves choosing a

    set of genes for analysis (e.g., kinases) and performing

    polymerase chain reaction (PCR) amplification of all

    coding exons including the intronexon splice site junctions

    for that set of genes, with genomic DNA isolated from

    patient tumor samples or cell lines serving as a template.

    The PCR is carried out using a high-fidelity polymerase to

    avoid introduction of de novo mutations during the

    amplification reaction. PCR products are purified and then

    subjected to bidirectional dideoxy sequencing to identify

    putative mutations. Ideally, patient-matched normal DNA is

    analyzed using the same procedure and then compared with

    results from sequencing of the tumor DNA to allow for

    distinction between possible mutations and natural poly-

    morphisms. This strategy for mutation identification has

    been successfully applied in studies of lung, colorectal,

    breast, and brain tumors [7, 27, 28, 122, 145, 146, 170,

    171]. For example, a resequencing project analyzing the

    receptor tyrosine kinase family of genes in human

    glioblastomas found mutations of the fibroblast growth

    receptor-1 gene and the platelet-derived growth factor

    receptor (PDGFR) gene [122]. To date, exon resequenc-

    ing has been most extensively applied to the tyrosine kinase

    and associated phosphatase gene families [7, 27, 28, 122,

    145, 146, 171]; however, there are no limitations on the

    types or families of genes that can be interrogated using this

    strategy.

    1382 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    5/16

    Molecular genetics of specific pediatric brain tumors

    High-grade astrocytoma

    High-grade astrocytomas in children are relatively rare

    lesions when compared with adults. Whereas the prognosis

    of children with anaplastic astrocytoma or glioblastoma

    multiforme may be better than that of adults with acomparable histological lesion, the majority of children

    will still succumb from progressive disease within 25 years

    after diagnosis [152]. What has been very interesting, and

    as yet incompletely characterized, is the fact that the genetic

    alterations that accompany the childhood high-grade astro-

    cytic neoplasms are distinct from those that occur in adults.

    The genetic alterations that are the hallmark of adult

    anaplastic astrocytomasloss of p53, PTEN, p14ARF, and

    amplification of the epidermal growth factor receptor

    (EGFR) III mutantare relatively rare in pediatric anaplas-

    tic astrocytomas (see below). In light of this and other

    evidence, it appears as if the development of pediatric high-grade astrocytomas may follow pathways distinct from the

    well-established primary and secondary paradigm of adult

    glioblastomas.

    Common chromosomal losses in pediatric high-grade

    gliomas include those of chromosome 16p, 17p, 19p, 19q,

    and 22 [59, 129]. Within the spectrum of high-grade

    gliomas, distinct cytogenetic changes are observed in

    pediatric anaplastic astrocytomas, which are typified by

    gains of chromosome 5q and losses of chromosomes 6q,

    9q, 12q, and 22q, and in pediatric glioblastoma, which is

    characterized by gains in chromosomes 1q and 16p and

    losses of chromosomes 8q and 17p [129]. The finding of 1q

    amplification is noteworthy because it may be a marker of

    poor survival.

    Whereas microsatellite instability appears to be generally

    absent in the setting of adult high-grade astrocytomas,

    nearly 25% of pediatric malignant astrocytomas may

    display a microsatellite instability phenotype as a manifes-

    tation of mutations in various DNA mismatch repair genes

    [1, 22].

    In addition to being characterized by their respective

    patterns of structural chromosomal abnormalities, pediatric

    high-grade astrocytomas may be distinguished from their

    adult counterparts on the basis of differential expression of

    oncogenes and tumor suppressor genes that are involved in

    signal transduction pathways critical to the process of

    gliomagenesis. The pertinent pathways for the adult tumors

    include the growth factor/growth factor receptor/PI3-kinase/

    Akt/PTEN pathway, the p53/MDM2/p14ARF pathway, and

    the pRB/cyclinD1/cyclin-dependent kinase (CDK) 4/p16

    pathway. The interested reader is referred to a review by

    Ichimura et al. for a recent synopsis of the interplay of these

    various pathways in the molecular pathogenesis of astro-

    cytic tumors [69]. In the remainder of this section, each

    pathway will be examined in turn in order to characterize

    the patterns of abnormality present within and to highlight

    the often dichotomous nature of the importance of an

    individual pathway to the genesis of pediatric vs adult high-

    grade gliomas.

    Although amplification of the EGFR gene is observed in

    up to 40% of adult glioblastomas and 15% of adultanaplastic astrocytomas, it is not a common finding in

    pediatric high-grade astrocytomas [22, 121, 148]. Similarly,

    whereas de novo adult glioblastomas have a high frequency

    of mutations in the PTEN tumor suppressor gene, pediatric

    malignant gliomas rarely contain such mutations. Although

    LOH at 10q2325 may be present in as many as 80% of

    informative cases, homozygous deletions of the PTEN gene

    are seen in only 8% of pediatric high-grade astrocytomas

    [22]. Where mutations of PTEN are observed, they may

    herald a poor prognosis for children with high-grade

    astrocytomas.

    The majority of adult secondary glioblastomas demon-strate mutations of the p53 tumor suppressor gene located

    on chromosome 17p. A relatively small subset of high-

    grade gliomas, mostly occurring in older children, may also

    harbor frequent p53 mutations. In this population, there

    appears to be a nearly 2:1 frequency of p53 gene mutations

    in high-grade brainstem astrocytomas as opposed to non-

    brainstem astrocytomas [22, 90, 117].

    Homozygous deletions of the CDKN2A/pl4ARF locus at

    9p21, which encodes both the p16 and p14ARFproteins, are

    found in 10% of pediatric malignant astrocytomas [103].

    Overexpression of the MDM2 oncogene is seen in 67% of

    pediatric malignant astrocytomas, although there is no

    concomitant amplification of the MDM2 gene at a genomic

    level [148]. Overall, inactivation of the p53/MDM2/p14ARF

    pathway, either by mutation of p53, overexpression of

    MDM2, or deletion of p14ARF, is seen in more than 95% of

    pediatric malignant astrocytomas, a situation which is

    similar to the observed frequency of p53/MDM2/p14ARF

    pathway inactivation in the setting of adult malignant

    astrocytomas [148]. However, as outlined earlier, mutations

    of p53 are seldom seen in malignant gliomas of children

    younger than 3 years, once again suggesting that malignant

    gliomas in very young children may follow a distinct

    molecular pathway as compared with older children and

    adults [118].

    Sure et al. were able to demonstrate loss of expression of

    p16 in 11 of 18 pediatric glioblastomas [149]. However, the

    pRb/cyclin D1/CDK4/p16 pathway appears to be inacti-

    vated in only about 25% of pediatric malignant astrocyto-

    mas, as opposed to inactivation in more than 80% of the

    corresponding adult tumors [148].

    To date, there have been no published reports of cDNA

    microarray analyses on pediatric high-grade gliomas.

    Childs Nerv Syst (2006) 22:13791394 1383

  • 8/9/2019 Child CNS

    6/16

    Low-grade astrocytoma

    Whereas tremendous advances have been made in the

    understanding of the molecular pathogenesis of pediatric

    high-grade astrocytomas, there have been comparatively

    few studies focusing on the low-grade and pilocytic

    astrocytomas of childhood.

    Consistent with the fact that they are low-grade lesions,cytogenetic studies have revealed a normal karyotype in the

    majority of pilocytic astrocytomas examined, and where

    abnormal profiles have been observed, no consistent

    karyotype abnormalities have been identified [138, 178].

    Pediatric pilocytic astrocytomas show fewer chromosomal

    changes than adult tumors; where observed, they are

    usually gains of only a single chromosome, such as

    chromosome 7 or 8, as has been demonstrated in

    approximately one third of pediatric pilocytic astrocytomas

    using fluorescent in situ hybridization (FISH) analysis

    [173].

    Individuals with neurofibromatosis type I (NF1) have anincreased propensity to develop pilocytic astrocytomas,

    especially of the optic/hypothalamic region. As sporadic

    pilocytic astrocytomas occasionally show LOH on chro-

    mosome 17q (the location of the NF1 tumor suppressor

    gene), one would predict that mutations of the NF1 gene

    with consequent loss of expression would be found in

    sporadic pilocytic astrocytomas. In fact, this appears not to

    be the case. In actuality, the expression of the NF1 tumor

    suppressor gene in sporadic pilocytic astrocytomas is often

    upregulated, perhaps as a reactive response to excessive

    cellular proliferation [116, 175]. This is in contrast to

    pilocytic astrocytomas arising in NF1 patients, where loss

    of NF1 expression is an obligatory finding [54]. The precise

    origin of the differing contribution of the NF1 gene product

    in the setting of NF1 vs non-NF1 pilocytic astrocytomas

    remains to be elucidated.

    Even with the use of various sensitive p53 mutation

    detection assays, it appears that p53 mutations are absent or

    at most infrequently present in the setting of pediatric

    pilocytic astrocytomas [23, 71, 174]. Such results are in

    concordance with the low frequency of allelic loss detected

    at the p53 locus on chromosome 17p [23, 71, 174]. Taken

    together, these findings indicate that abnormalities of p53

    do not contribute in any significant way to the genesis of

    pilocytic astrocytomas.

    Most cases of pediatric pilocytic astrocytoma show

    immunopositivity for p16 and CDK4, indicating that

    abnormalities in the pRb/cyclinD1/CDK4/p16 pathway

    likely do not play an important role in the evolution of

    these tumors [23]. PTEN mutations are also likely not a

    significant contributor to the pathogenesis of these lesions

    [23, 37], as mutations in this tumor suppressor gene tend to

    be reserved for higher-grade tumors [123].

    Ependymoma

    Chromosomal 22 defects are frequently found in ependy-

    moma. Mutation of the NF-2 gene product on chromosome

    22 has been documented to predispose to the formation of

    various tumor types, including ependymoma, especially in

    patients with NF-2 [41]. However, the vast majority of

    sporadic (non-NF-2) cases lack mutations in the NF-2 gene.The most plausible explanation for these findings is the

    existence of another oncogene or tumor suppressor gene on

    chromosome 22 that is more commonly involved in the

    genesis of sporadic ependymoma than the NF-2 gene

    product [166].

    Pediatric intracranial ependymomas appear to have a

    chromosomal signature distinct from their adult counter-

    parts. Pediatric tumors, which often occur in the

    infratentorial compartment, display balanced CGH pro-

    files in 3050% of cases, in comparison to only 10% of

    adult tumors [38, 63]. This finding suggests that pediatric

    intracranial ependymomas may progress along substantiallydifferent pathways than those giving rise to adult supra-

    tentorial or spinal ependymomas [161].

    Where chromosomal aberrations are observed in pediat-

    ric ependymomas, monosomy 17 appears to be one of the

    most common lesions, with an approximately 50% fre-

    quency [166]. The p53 tumor suppressor gene resides on

    17p, and individual case reports have identified some

    instances of p53 mutation in ependymoma patients,

    including a family who had concurrent mutation in

    chromosome 22 [46, 104].

    Gain of chromosome 1q is also a frequent finding in

    pediatric ependymoma [38]. A putative region of interest on

    chromosome 1q may be the region between 1q22 and 31

    [82]. Several studies have made an association between the

    presence of 1q gain and a poor clinical course, suggesting

    that the presence of one or more genes located on 1q may

    be responsible for tumor progression and/or response to

    therapy [38, 63].

    Loss of a region on 6q has been documented in a number

    of case reports; however, no known oncogene or tumor

    suppressor gene has yet been implicated at this locus [82,

    125, 131]. Other frequent chromosomal alterations include

    losses of chromosomes 1p, 6, 9q, 11, 13q, 16p, 16q, 19q,

    20p, and 20q, and gains of 1q, 2, 5, 9, 12, 15, 18, 20q, and

    X [63, 163].

    Microarray experiments designed to document the gene

    expression profile of ependymomas have been performed.

    A recent examination of a panel of 12,627 genes identified

    a subset of 112 genes as being abnormally expressed in

    pediatric ependymoma when compared with normal brain

    controls [147]. Genes with increased expression included

    the oncogene WNT5A, the p53 homologue p63, and

    several cell cycle, cell adhesion, and cell proliferation

    1384 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    7/16

    genes. Underexpressed genes included the NF2 interacting

    gene SCHIP-1 and the adenomatous polyposis coli (APC)-

    associated gene EB1 among others. These genes represent

    candidate genes for further study; further validation work is

    necessary in order to clarify their precise contribution to

    ependymoma tumorigenesis.

    Our understanding of the origins of ependymoma

    increased dramatically with the recent publication of Tayloret al. in which histologically identical, but genetically

    distinct, ependymomas showed patterns of gene expression

    that recapitulate those of radial glia cells in corresponding

    regions of the central nervous system [161]. In this study,

    supratentorial, infratentorial, and intraspinal ependymomas

    demonstrated distinct genetic signatures and were shown to

    arise from restricted populations of radial glia stem cells.

    For the supratentorial tumors, CDK4 and several Notch

    signaling pathway genes were overexpressed; for the

    infratentorial tumors, IFG-1 and several HOX homeobox

    genes were overexpressed; and for the spinal tumors, the ID

    genes and the aquaporins were overexpressed. The impli-cations of this study are that ependymomas should be

    treated with therapies that target the cell signal pathways

    that maintain subsets of ependymoma stem cells rather than

    the histological or clinical forms of the disease [161].

    Atypical teratoid rhabdoid tumor

    Molecular studies have been able to distinguish CNS AT/RT

    from the other primitive pediatric brain tumors, establishing

    it as a unique pathological entity [55, 133, 134]. AT/RT

    frequently demonstrates deletion of the long arm of

    chromosome 22q11.2, and further molecular studies have

    led to the identification of the INI1/hSNF5 tumor suppres-

    sor gene at this location [1013, 165]. A somatic mutation

    in this gene predisposes children to develop AT/RT

    [1013]. The hSNF5 protein is the smallest member of a

    highly conserved family of proteins that function in

    chromatin remodeling via the nucleosome. By winding

    and unwinding DNA, this complex changes the config-

    uration of genomic DNA, thus allowing or denying

    transcription factors access to the DNA, consequently

    changing patterns of gene expression.

    Some children with AT/RT are born with heterozygous

    germ line mutations of the hSNF5 gene, suggesting that

    these children were predisposed to develop AT/RT [155]; in

    most cases, however, these germ line mutations arise de

    novo.

    Heterozygous mSNF5 +/ knockout mice develop

    tumors resembling AT/RT, supporting the role of hSNF5 as

    a tumor suppressor gene [130]. Although most AT/RTs

    show evidence of some genetic derangement at the hSNF5

    locus, mutational analysis of the hSNF5 gene in a series of

    primitive neuroectodermal tumors/medulloblastomas dis-

    covered mutations in only 4 of 52 tumors [155]. Of those

    four, two were reclassified as AT/RT upon reexamination of

    the pathology, but there was insufficient clinical material to

    establish an accurate diagnosis in the other two cases. This

    suggests that tumors which are histologically diagnosed as

    primitive neuroectodermal tumors/medulloblastomas but

    which also harbor hSNF5 mutations are most likely AT/RT.

    Although mutation/deletion of hSNF5 is not currentlysufficient for a diagnosis of AT/RT, it appears to be related

    to the clinical outcome; consequently, determination of the

    status of the AT/RT gene by DNA sequence analysis, FISH,

    or immunohistochemistry is rapidly becoming an integral

    part of the neuropathological diagnosis of primitive

    pediatric malignant brain tumors (Fig. 2).

    Medulloblastoma

    Recent gene expression studies have shown that medullo-

    blastoma is molecularly distinct from the supratentorial

    primitive neuroectodermal tumors (sPNETs) [119]. TheSonic Hedgehog (SHH), Wingless(WNT/WG), and

    receptor tyrosine kinase I family ERBB pathway are

    emerging as central developmental signaling pathway

    systems in the formation of medulloblastoma [17, 110].

    The specific contributions of these pathways to the genesis

    of medulloblastoma will be discussed in turn.

    Medulloblastoma has been reported in the setting of a

    number of different hereditary cancer syndromes, including

    nevoid basal cell carcinoma syndrome (NBCCS), Turcot

    syndrome, LiFraumeni syndrome and RubensteinTaybi

    syndrome [156]. In the mid-1990s, it was discovered that

    NBCCS arises from germ line mutations of PTCH1, which

    Fig. 2 Pattern of INI-1 staining in AT/RTs of childhood. Immuno-

    histochemical staining with an anti-INI-1 antibody fails to stain the

    nuclei of tumor cells but does stain the nuclei of normal or reactive

    tissues in the vicinity

    Childs Nerv Syst (2006) 22:13791394 1385

  • 8/9/2019 Child CNS

    8/16

    encodes the receptor for the Hedgehog (Hh) family of

    signaling proteins. PTCH1 is a 12-pass transmembrane

    receptor protein that represses activity of the Hh signaling

    pathway in its unbound state by suppressing the activity of

    the seven-pass transmembrane protein Smoothened

    (SMOH) [56, 74, 94]. Upon ligand binding, PTCH1s tonic

    inhibition of SMOH is lifted, leading to the release of GLI

    transcription factor from a tetra-protein complex which alsoincludes suppressor of fused (SUFU), fused (FU), and

    costal-2 (COS2). Upon its release, GLI is activated and

    translocated to the nucleus where it binds to promoters in a

    sequence-specific fashion to increase the transcription of

    various growth-promoting genes such as cyclin D [94].

    Mutations in the various molecules comprising this signal-

    ing complex, including SHH, PTCH1, SMOH, and SUFU,

    have been shown to occur in both familial and sporadic

    tumors [43, 113, 126, 158]. Another recent report demon-

    strated a crucial role for polycomb group gene Bmi1 in

    clonal expansion of granule cell precursors in vivo and

    linked overexpression of BMI1 and patched (PTCH), afinding that is suggestive of an alternative or additive

    mechanism of SHH pathway activation in the pathogenesis

    of medulloblastomas [84]. A novel putative tumor suppres-

    sor, human REN(KCTD11), incidentally maps to 17p13.2,

    a chromosomal region which is frequently lost in medul-

    loblastoma [45]. The REN tumor suppressor inhibits

    medulloblastoma growth by impairing both Gli2-dependent

    gene transcription and SHH-enhanced expression of the

    target Gli1 mRNA, thereby decreasing the expression of

    downstream genes [34, 35].

    Cyclopamine, a plant-derived teratogen that binds to

    SMOH and interrupts its ability to activate downstream

    signaling, may have some important therapeutic implica-

    tions. Mice harboring human medulloblastoma explants

    showed tumor regression upon drug administration. Less

    toxic derivatives of cyclopamine are being used in

    preclinical trials of medulloblastoma. Although this com-

    pound will probably only be effective for patients with

    NBCCS or those whose tumors harbor PTCH and SMOH

    mutations, it carries great potential as a targeted molecular

    therapy in such instances [9, 151].

    Germ line mutations of APC in patients with Turcot

    syndrome have been reported to increase the risk of

    developing medulloblastomas [58]. The inactivation of the

    APC gene leads to aberrant signaling in the WNT pathway,

    resulting in the inability of the multiprotein complex

    containing APC, axin, and glycogen synthase kinase-3

    (GSK-3) to phosphorylate and cause degradation of -

    catenin. This leads to an overabundance of -catenin and

    its translocation to the nucleus where it activates the TCF/

    LEF transcriptional complex. The -catenin/TCF dimer

    activates the transcription of other growth-regulating genes,

    some of which are known oncogenes (e.g., c-MYC and

    cyclin D) [42, 101, 150]. Mutations in WNT pathway

    members occur in about 15% of sporadic medulloblastoma

    [25, 66, 77, 181]. A recent report also demonstrated that

    mutations in SUFU can cause a decreased efficiency of-

    catenin nuclear export, thereby also resulting in increased

    -catenin/TCF signaling [160].

    The ERBB or epidermal growth factor family of receptor

    tyrosine kinases plays a crucial role in regulating cellular proliferation, apoptosis, migration, and differentiation.

    Activation of these receptors through ligand binding,

    dimerization, and autophosphorylation culminate in down-

    stream signaling through mitogen-activated protein kinase

    (MAPK), AKT, and STAT [107]. Interestingly, one of the

    four members of the ERBB family, ERBB2, has been

    shown to be highly expressed in medulloblastoma [52]. A

    high level of ERBB2 and ERBB4 coexpression signifies an

    increased risk of metastases and is associated with poor

    prognosis in cases of medulloblastoma [48, 50]. Microarray

    analysis of medulloblastoma cell lines demonstrate that

    S100A4, a prometastatic gene known to be linked to breastand bladder cancers, is a direct target of ERBB2 signaling

    in medulloblastoma cells via the AKT/PI3K pathway [61].

    Compounds such as OSI-774 (erlotinib, also known as

    gefitinif) inhibit ERBB2 signaling in human medulloblas-

    toma cells and may have therapeutic potential [61].

    The PDGFR and downstream activation of the RAS/

    MAPK signaling pathway (including MAP2K1, MAP2K2,

    and MAPK1/3) have also been uncovered as potential

    mediators of medulloblastoma metastasis [33, 51, 92].

    The mRNA expression level of TrkC, a neurotrophin

    signaling receptor, was identified as a positive prognostic

    factor for progression-free and overall survival in medul-

    loblastoma [53, 124]. The MYC protooncogenes (MYCN

    and MYCC) are amplified and/or overexpressed in a

    subset of large cell medulloblastomas and also correlate

    with poor clinical outcome [39, 40]. However, other

    studies of TrkC, MYCC, and MYCN mRNA expression

    or immunoreactivity did not demonstrate a significant

    prognostic effect [48, 80].

    Loss of chromosome 17p, often through the formation of

    an isochromosome 17q i(17)(q10), is the most common

    chromosome aberration in childhood medulloblastoma,

    occurring in about 25% to 35% of cases. Either isolated

    17p deletion or i(17)(q10) has been reported as a significant

    negative prognostic factor [50, 109]. Recent cytogenetic

    analysis using matrix CGH suggested that overexpression

    of CDK6 correlates with a worse prognosis [99].

    Most of the tumor suppressor genes shown to play a role

    in the formation of medulloblastoma have been identified

    through mutations; however, other epigenetic phenomena

    may lead to their decreased expression. Aberrant methyla-

    tion of CpG islands located in promoter regions represents

    one of the major mechanisms for silencing of cancer-related

    1386 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    9/16

    genes in tumor cells. Extensive hypermethylation of the

    RASSF1A gene (ras-association domain family protein 1,

    isoform A), an identified tumor suppressor gene located at

    chromosome 3p21.3, occurs in about 80% of primary

    medulloblastomas [86, 91]. Furthermore, complete methyl-

    ation of the putative tumor suppressor HIC-1 (hyper-

    methylated in cancer) and the apoptosis effector molecule

    caspsase-8 has been found in a subset of medulloblastoma[132, 168, 182].

    Germ cell tumors

    Although the exact molecular and cytogenetic aberrations

    in GCTs are still not well defined, the most consistent

    cytogenetic abnormality observed in such tumors is

    isochromosome 12p (i12p) [26, 31, 89]. Some tumors

    without i12p have overrepresentation of 12p by other

    mechanisms, such as duplication of the entire chromosome.

    The exact role of genes implicated in 12p overrepresenta-tion in GCTs remains uncertain, but evidence suggests that

    the genes located on this chromosome, such as cyclin D2,

    may play a role in facilitating entry into S phase of the cell

    cycle. The rather frequent finding of i12p is also highly

    indicative of the existence of novel putative oncogenes on

    12p which may be involved in the pathogenesis of GCTs.

    Other chromosomal aberrations have also been observed

    in pineal GCTs. In a study of 15 pineal region GCTs,

    Rickert et al. reported that the most common chromosomal

    imbalances in pineal germinomas were losses of 13q and

    18q, and loss of chromosomes 4 and 5 [128]. Interestingly,

    these authors infrequently found gain of 12p. A summary

    of other reported genetic aberrations in these tumors can be

    found in a recent review by Taylor et al. [157].

    Although most GCTs are sporadic, a few genetic

    syndromes do predispose individuals to their development.

    Pineal germinomas or teratomas in patients with Klinefelter

    syndrome have been reported [120]. That patients with

    Klinefelter syndrome generally have increased risk of

    developing malignancy is in keeping with the idea of the

    existence of a putative oncogene on the X chromosome.

    Patients with trisomy 21 have an increased risk of a number

    of cancers including leukemia and gonadal and extragona-

    dal GCTs [139].

    Ewing sarcoma

    Primary Ewing sarcoma (ES)/peripheral primitive neuro-

    ectodermal tumors (pPNET) of the central nervous system

    constitute a clinically important, albeit rare, subset of

    pediatric soft tissue tumors. They typically arise extracra-

    nially or paraspinally and often result in secondary invasion

    of critical neural structures [18].

    In recent years, an examination of the genetic alterations

    present within the spectrum of pediatric soft tissue tumors

    has demonstrated a fairly specific (although not absolute)

    association between specific nonrandom reciprocal chro-

    mosomal translocations and individual soft tissue tumor

    types [20]. Ongoing study of these fusion products has

    yielded profound insights into the biology of these tumors

    and may hold great promise for novel diagnostic andtherapeutic applications.

    Ewing sarcoma/pPNET-associated translocations charac-

    teristically involve the EWS gene on 22q12 and various

    members of the ETS family of protooncogenes (Fig. 3).

    Approximately 85% of ES/pPNET tumors harbor the

    translocation t(11;22)(q24;q12); in this subset of tumors,

    the translocation partner for EWS is the FLI1 gene product

    found on 11q24 [32]. In nearly 10% of cases, the

    translocation partner is ERG [t(21;22)(q22;q12)] [144],

    and in rare cases, EWS may be fused to the ETS domains

    of ETV-1 [t(7;22)(p22;q12)] [73], E1AF [t(17;22)(q12;

    q12)] [76], or FEV [t(2;22)(q33;q12)] [111].The EWS gene product is a member of a growing family

    of highly conserved RNA-binding proteins [105]. Although

    the exact biological function of wild-type EWS and its

    homologues remains largely unknown, a growing body of

    evidence suggests that they are involved in mRNA

    transcription. EWS has been shown to form an adaptor

    ternary complex with RNA polymerase II and other

    heterogeneous RNA-binding proteins [112]; these findings

    are highly suggestive of an important role of EWS in basic

    transcriptional regulation.

    ETS-domain-containing proteins are DNA-binding tran-

    scription factors that are implicated in the control of cellular

    proliferation. ETS family members appear to cooperate

    with other nuclear proteins to help establish promoter

    specificity, modulate transcriptional regulation, and facili-

    tate linkage to various signal transduction pathways,

    including RAS [36, 64, 143, 172].

    The ES/pPNET-associated translocations result in chi-

    meric proteins containing the N-terminal domain of EWS

    fused to the site-specific nucleic acid binding domain of the

    ETS transcription factor translocation partner (Fig. 3). This

    structure suggests that the chimeric protein is directed at the

    promoter region of specific genes recognized by the

    translocated DNA-binding domain of the ETS member [5,

    106]. However, the ultimate targets of the EWS-ETS

    chimeric protein may not be solely dependent on the ETS

    domain; rather, other proteinprotein interactions unique to

    the chimeric molecule may be at play [98]. The actual

    target genes contributing to tumorigenesis are not known,

    but analysis of mRNAs differentially expressed in cell lines

    stably transfected with the EWS-FLI1 fusion has produced

    some interesting candidates, including manic fringe, c-myc,

    cyclin D1, mE2-C, MMP-1, and TGF-RIII [4, 6, 14].

    Childs Nerv Syst (2006) 22:13791394 1387

  • 8/9/2019 Child CNS

    10/16

    The fairly consistent presence of these recombinant gene

    products in ES/pPNET suggests that they play a critical role

    in the underlying biology of these tumors (Fig. 3). Trans-fection of EWS-FLI1 or EWS-ERG can transform mouse

    NIH-3T3 cells if both the EWS and ETS domains are

    functionally intact [98]. EWS-FLI1 antisense RNA trans-

    fected into ES/pPNET cells results in marked growth

    inhibition, suggesting that the EWS-ETS gene rearrange-

    ment may also be necessary for maintaining the malignant

    phenotype of ES/pPNET cell lines [153]. Furthermore,

    expression of EWS-ETS fusion constructs may contribute

    to tumorigenesis via inhibition of apoptosis; not surprisingly,

    antisense inhibition of EWS-ETS fusion genes may enhance

    susceptibility to chemotherapy-induced apoptosis [177].

    The gene fusions characteristic of ES/pPNET exhibit an

    underlying molecular heterogeneity. There are two under-

    lying sources of variability: the specific ETS fusion partner

    and the breakpoint location within the genes. A better

    outcome for patients with localized tumors expressing the

    most common chimeric transcript (type I: EWS exon 7

    fused to FLI1 exon 6) compared with the next most

    common fusion types (type II: EWS exon 7 fused to FLI1

    exon 5 or type III: EWS exon 10 fused to FLI1 exon 6) has

    been reported, raising the possibility that heterogeneity in

    chimeric transcripts may reliably define clinically distinct

    risk groups [30, 85]. Preliminary work suggests that the

    better outcome associated with type I fusion transcripts maybe related to the weaker transcriptional activation properties

    of the type I transcript [30].

    Other less common numerical and structural chromo-

    somal aberrations may also be found in ES/pPNET [3, 15,

    97, 102, 108, 154]. The most common numerical abnor-

    malities are +1q, +8, +12, and +20, and 1p, 16q, and

    19q. More complex, multichromosome translocations,

    such as t(11;14;22)(q24;q11;q12) and t(10;11;22)(p11.2;

    q24;q12), may also occur and generally portend a poor

    prognosis.

    Changes in known tumor suppressor genes may be

    observed in some cases. Homozygous deletion of

    CDKN2A (p14ARF) on 9p21 has been described in

    approximately 30% of cases [81]. Although mutations in

    p53 have been described in up to 50% of ES/pPNET cell

    lines [57], this appears to be a rare event in primary tumors

    [167]. Amplification of the MDM2 gene, an inactivator of

    p53, is also rare in ES/pPNET, consistent with the

    hypothesis that p53 and regulators of its activity may not

    play a dominant role in the pathogenesis of ES/pPNET

    [93].

    Fig. 3 Schematic representation

    of the two commonest fusion

    genes in Ewing sarcoma and the

    proposed effects of the onco-

    genic chimeric protein on cellu-

    lar proliferation and

    tumorigenicity

    1388 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    11/16

    Conclusions

    As is evident from this review, our understanding of the

    fundamental mechanisms of brain tumorigenesis in children

    has increased markedly over the past 2 decades. The pace

    of subsequent advances will only quicken, owing to such

    monumental technical feats as the sequencing of the human

    genome. Concomitant developments in the field of bio-informatics will be equally important as they will allow us

    to make sense of the volumes of data that will undoubtedly

    emerge from the ongoing interrogation of the fundamental

    molecular processes at work in the genesis of pediatric

    brain tumors. The challenge for the next 2 decades will be

    to consolidate our understanding of the molecular patho-

    genesis of childhood brain tumors and to apply this

    sophisticated knowledge toward the development of clini-

    cally useful treatment strategies.

    Acknowledgements This work was supported by grants from the

    National Cancer Institute of Canada, the Pediatric Brain Tumor

    Foundation, the Wiley Fund, and the Laurie Berman Fund for BrainTumor Research.

    References

    1. Alonso M, Hamelin R, Kim M, Porwancher K, Sung T, Parhar P,

    Miller DC, Newcomb EW (2001) Microsatellite instability

    occurs in distinct subtypes of pediatric but not adult central

    nervous system tumors. Cancer Res 61:21242128

    2. Altshuler D, Brooks LD, Chakravarti A, Collins FS, Daly MJ,

    Donnelly P, International HapMap Consortium (2005) A

    haplotype map of the human genome. Nature 437:12991320

    3. Armengol G, Tarkkanen M, Virolainen M et al (1997) Recurrent

    gains of 1q, 8 and 12 in the Ewing family of tumors by

    comparative genomic hybridization. Br J Cancer 75:14031409

    4. Baer C, Nees M, Breit S et al (2004) Profiling and functional

    annotation of mRNA gene expression in pediatric rhabdomyo-

    sarcoma and Ewings sarcoma. Int J Cancer 110:687694

    5. Bailly RA, Bosselut R, Zucman J et al (1994) DNA-binding and

    transcriptional activation properties of the EWS-FLI-1 fusion

    protein resulting from the t(11;22) translocation in Ewing

    sarcoma. Mol Cell Biol 14:32303241

    6. Bandres E, Malumbres R, Escalada A et al (2005) Gene

    expression profile of Ewing sarcoma cell lines differing in their

    EWS-FLI1 fusion type. J Pediatr Hematol Oncol 27:537542

    7. Bardelli A, Parsons DW, Silliman N, Ptak J, Szabo S, Saha S,

    Markowitz S, Willson JK, Parmigiani G, Kinzler KW, Vogelstein

    B, Velculescu VE (2003) Mutational analysis of the tyrosinekinome in colorectal cancers. Science 300:949

    8. Barrett MT, Scheffer A, Ben-Dor A, Sampas N, Lipson D,

    Kincaid R, Tsang P, Curry B, Baird K, Meltzer PS, Yakhini Z,

    Bruhn L, Laderman S (2004) Comparative genomic hybridiza-

    tion using oligonucleotide microarrays and total genomic DNA.

    Proc Natl Acad Sci USA 101:1776517770

    9. Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI,

    Eberhart CG, Watkins DN et al (2002) Medulloblastoma

    growth inhibition by hedgehog pathway blockade. Science

    297:15591561

    10. Biegel JA, Rorke LB, Packer RJ et al (1990) Monosomy 22 in

    rhabdoid or atypical tumors of the brain. J Neurosurg 73:710 714

    11. Biegel JA, Zhou JY, Rorke LB et al (1999) Germ-line and

    acquired mutations of INI1 in atypical teratoid and rhabdoid

    tumors. Cancer Res 59:7479

    12. Biegel JA, Fogelgren B, Zhou JY, James CD, Janss AJ, Allen JC,

    Zagzag D, Raffel C, Rorke LB (2000) Mutations of the INI1

    rhabdoid tumor suppressor gene in medulloblastomas and

    primitive neuroectodermal tumors of the central nervous system.

    Clin Cancer Res 6:27592763

    13. Biegel JA, Tan L, Zhang F, Wainwright L, Russo P, Rorke LB

    (2002) Alterations of the hSNF5/INI1 gene in central nervous

    system atypical teratoid/rhabdoid tumors and renal and extra-

    renal rhabdoid tumors. Clin Cancer Res 8:34613467

    14. Braun BS, Frieden R, Lessnick SL et al (1995) Identification

    of target genes for the Ewings sarcoma EWS/Fli fusion

    protein by representational difference analysis. Mol Cell Biol

    15:46234630

    15. Brisset S, Schleiermacher G, Peter M et al (2001) CGH analysis

    of secondary genetic changes in Ewing tumors: correlation with

    metastatic disease in a series of 43 cases. Cancer Genet

    Cytogenet 130:5761

    16. Bucca G, Carruba G, Saetta A, Muti P, Castagnetta L, Smith CP

    (2004) Gene expression profiling of human cancers. Ann N Y

    Acad Sci 1028:2837

    17. Buhren J, Christoph AH, Buslei R, Albrecht S, Wiestler OD,

    Pietsch T (2000) Expression of the neurotrophin receptor

    p75NTR in medulloblastomas is correlated with distinct

    histological and clinical features: evidence for a medulloblasto-

    ma subtype derived from the external granule cell layer. J

    Neuropathol Exp Neurol 59:229240

    18. Carlotti CG Jr, Drake JM, Hladky JP, Teshima I, Becker LE,

    Rutka JT (1999) Primary Ewings sarcoma of the skull in

    children. Utility of molecular diagnostics, surgery and adjuvant

    therapies. Pediatr Neurosurg 31:307315

    19. Carvalho B, Ouwerkerk E, Meijer GA, Ylstra B (2004) High

    resolution microarray comparative genomic hybridisation analy-

    sis using spotted oligonucleotides. J Clin Pathol 57:644646

    20. Chang CC, Shidham VB (2003) Molecular genetics of pediatric

    soft tissue tumors. J Mol Diagn 5:143154

    21. Cheng J, Kapranov P, Drenkow J, Dike S, Brubaker S, Patel S,

    Long J, Stern D, Tammana H, Helt G, Sementchenko V,

    Piccolboni A, Bekiranov S, Bailey DK, Ganesh M, Ghosh S,

    Bell I, Gerhard DS, Gingeras TR (2005) Transcriptional maps of

    10 human chromosomes at 5-nucleotide resolution. Science

    308:11491154

    22. Cheng Y, Ng HK, Zhang SF, Ding M, Pang JC, Zheng J, Poon

    WS (1999) Genetic alterations in pediatric high-grade astrocyto-

    mas. Hum Pathol 30:12841290

    23. Cheng Y, Pang JC, Ng HK, Ding M, Zhang SF, Zheng J, Liu

    DG, Poon WS (2000) Pilocytic astrocytomas do not show most

    of the genetic changes commonly seen in diffuse astrocytomas.

    Histopathology 37:437444

    24. Cowell JK, Matsui S, Wang YD, LaDuca J, Conroy J, McQuaid

    D, Nowak NJ (2004) Application of bacterial artificial chromo-

    some array-based comparative genomic hybridization and spec-tral karyotyping to the analysis of glioblastoma multiforme.

    Cancer Genet Cytogenet 151:3651

    25. Dahmen RP, Koch A, Denkhaus D et al (2001) Deletions of

    AXIN1, a component of the WNT/wingless pathway, in sporadic

    medulloblastomas. Cancer Res 61:70397043

    26. Dal Cin P, Dei Tos AP, Qi H, Giannini C, Furlanetto A, Longatti

    PL, Marynen P, Van den Berghe H (1998) Immature teratoma of

    the pineal gland with isochromosome 12p. Acta Neuropathol

    (Berl) 95:107110

    27. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S,

    Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N,

    Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J,

    Childs Nerv Syst (2006) 22:13791394 1389

  • 8/9/2019 Child CNS

    12/16

    Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S,

    Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C,

    Shipley J, Hargrave D, Pritchard-Jones K, Maitland N,

    Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu

    A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber

    BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ,

    Wooster R, Stratton MR, Futreal PA (2002) Mutations of the

    BRAF gene in human cancer. Nature 417:949954

    28. Davies H, Hunter C, Smith R, Stephens P, Greenman C, Bignell

    G, Teague J, Butler A, Edkins S, Stevens C, Parker A, OMeara S,Avis T, Barthorpe S, Brackenbury L, Buck G, Clements J, Cole J,

    Dicks E, Edwards K, Forbes S, Gorton M, Gray K, Halliday K,

    Harrison R, Hills K, Hinton J, Jones D, Kosmidou V, Laman R,

    Lugg R, Menzies A, Perry J, Petty R, Raine K, Shepherd R, Small

    A, Solomon H, Stephens Y, Tofts C, Varian J, Webb A, West S,

    Widaa S, Yates A, Brasseur F, Cooper CS, Flanagan AM, Green

    A, Knowles M, Leung SY, Looijenga LH, Malkowicz B, Pierotti

    MA, Teh BT, Yuen ST, Lakhani SR, Easton DF, Weber BL,

    Goldstraw P, Nicholson AG, Wooster R, Stratton MR, Futreal PA

    (2005) Somatic mutations of the protein kinase gene family in

    human lung cancer. Cancer Res 65:75917595

    29. Davies JJ, Wilson IM, Lam WL (2005) Array CGH technologies

    and their applications to cancer genomes. Chromosome Res

    13:237248

    30. de Alava E, Panizo A, Antonescu CR, Huvos AG, Pardo-Mindan

    FJ, Barr FG, Ladanyi M (2000) Association of EWS-FLI1 type 1

    fusion with lower proliferative rate in Ewings sarcoma. Am J

    Pathol 156:849855

    31. de Bruin TW, Slater RM, Defferrari R, Geurts van Kessel A,

    Suijkerbuijk RF, Jansen G, de Jong B, Oosterhuis JW (1994)

    Isochromosome 12p-positive pineal germ cell tumor. Cancer Res

    54:15421544

    32. Delattre O, Zucman J, Plougastel B et al (1992) Gene fusion with

    an ETS DNA-binding domain caused by chromosome translo-

    cation in human tumors. Nature 359:162165

    33. Del Valle L, Enam S, Lassak A, Wang JY, Croul S et al (2002)

    Insulin-like growth factor I receptor activity in human medullo-

    blastomas. Clin Cancer Res 8:18221830

    34. De Smaele E, Di Marcotullio L, Ferretti E, Screpanti I, Alesse E,

    Gulino A (2004) Chromosome 17p deletion in human medullo-

    blastoma: a missing checkpoint in the Hedgehog pathway. Cell

    Cycle 3:12631266

    35. Di Marcotullio L, Ferretti E, De Smaele E, Argenti B, Mincione

    C et al (2004) REN(KCTD11) is a suppressor of Hedgehog

    signaling and is deleted in human medulloblastoma. Proc Natl

    Acad Sci USA 101:1083310838

    36. Dittmer J, Nordheim A (1998) ETS transcription factors and

    human disease. Biochim Biophys Acta 1377:F1F11

    37. Duerr EM, Rollbrocker B, Hayashi Y, Peters N, Meyer-Puttlitz

    B, Louis DN, Schramm J, Wiestler OD, Parsons R, Eng C, von

    Deimling A (1998) PTEN mutations in gliomas and glioneuronal

    tumors. Oncogene 16:22592264

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

    Grundy R (2002) Genomic imbalances in pediatric intracranial

    ependymomas define clinically relevant groups. Am J Pathol

    161:21332141

    39. Eberhart CG, Kepner JL, Goldthwaite PT et al (2002) Histo-

    pathologic grading of medulloblastomas: a Pediatric Oncology

    Group study. Cancer 94:552560

    40. Eberhart CG, Kratz J, Wang Y, Summers K, Stearns D et al

    (2004) Histopathological and molecular prognostic markers in

    medulloblastoma: c-myc, n-myc, TrkC, and anaplasia. J

    Neuropathol Exp Neurol 63:441449

    41. Ebert C, von Haken M, Meyer-Puttlitz B, Wiestler OD,

    Reifenberger G, Pietsch T, von Deimling A (1999) Molecular

    genetic analysis of ependymal tumors. NF2 mutations and

    chromosome 22q loss occur preferentially in intramedullary

    spinal ependymomas. Am J Pathol 155:627632

    42. Elberhart CG, Tihan T, Burger PC (2000) Nuclear localization

    and mutation of beta-catenin in medulloblastoma. J Neuropathol

    Exp Neurol 59:333337

    43. Erez A, Ilan T, Amariglio N et al (2002) GLI3 is not mutated

    commonly in sporadic medulloblastomas. Cancer 95:2831

    44. Fernandez-Teijeiro A, Betensky RA, Sturla LM, Kim JY,

    Tamayo P, Pomeroy SL (2004) Combining gene expression

    profiles and clinical parameters for risk stratification in medul-loblastomas. J Clin Oncol 22:994998

    45. Ferretti E, Smaele ED, Marcotullio LD, Screpanti I, Gulino A

    (2005) Hedgehog checkpoints in medulloblastoma: the chromo-

    some 17p deletion paradigm. Trends Mol Med 11:537545

    46. Fink KL, Rushing EJ, Schold SC Jr, Nisen PD (1996)

    Infrequency of p53 gene mutations in ependymomas. J Neuro-

    oncol 27:111115

    47. Freeman WM, Robertson DJ, Vrana KE (2000) Fundamentals of

    DNA hybridization arrays for gene expression analysis. Bio-

    techniques 29:10421046, 10481055

    48. Gajjar A, Hernan R, Kocak M, Fuller C, Lee Y et al (2004)

    Clinical, histopathological, and molecular markers of prognosis:

    toward a new disease risk stratification system for medulloblas-

    toma. J Clin Oncol 22:984993

    49. Garraway LA, Widlund HR, Rubin MA, Getz G, Berger AJ,

    Ramaswamy S, Beroukhim R, Milner DA, Granter SR, Du J,

    Lee C, Wagner SN, Li C, Golub TR, Rimm DL, Meyerson ML,

    Fisher DE, Sellers WR (2005) Integrative genomic analyses

    identify MITF as a lineage survival oncogene amplified in

    malignant melanoma. Nature 436:117122

    50. Gilbertson R, Wickramasinghe C, Hernan R, Balaji V, Hunt D et

    al (2001) Clinical and molecular stratification of disease risk in

    medulloblastoma. Br J Cancer 85:705712

    51. Gilbertson RJ, Clifford SC (2003) PDGFRB is overexpressed in

    metastatic medulloblastoma. Nat Genet 35:197198

    52. Gilbertson RJ, Clifford SC, MacMeekin W, Meekin W, Wright C

    et al (1998) Expression of the ErbB-neuregulin signaling network

    during human cerebellar development: implications for the

    biology of medulloblastoma. Cancer Res 58:39323941

    53. Grotzer MA, Janss AJ, Fung KM, Biegel JA, Sutton LN, Rorke

    LB (2000) TrkC expression predicts good clinical outcome in

    primitive neuroectodermal brain tumors. J Clin Oncol 18:1027

    1035

    54. Gutmann DH, Donahoe J, Brown T, James CD, Perry A (2000)

    Loss of neurofibromatosis 1 (NF1) gene expression in NF1-

    associated pilocytic astrocytomas. Neuropathol Appl Neurobiol

    26:361367

    55. Gyure KA (2005) Newly defined central nervous system

    neoplasms. Am J Clin Pathol 123(Suppl):S3S12

    56. Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S

    et al (1996) Mutations of the human homolog of Drosophila

    patched in the nevoid basal cell carcinoma syndrome. Cell

    85:841851

    57. Hamelin R, Zucman J, Melot T et al (1994) p53 mutations in

    human tumors with chimeric EWS/FLI-1 genes. Int J Cancer

    57:336340

    58. Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell

    SM et al (1995) The molecular basis of Turcots syndrome. N

    Engl J Med 332:839847

    59. Harkness W, Hayward R, Darling J, Thomas D (2001)

    Identification of extensive genomic loss and gain by comparative

    genomic hybridisation in malignant astrocytoma in children and

    young adults. Genes Chromosomes Cancer 31:1522

    60. Harrington CA, Rosenow C, Retief J (2000) Monitoring gene

    expression using DNA microarrays. Curr Opin Microbiol

    3:285291

    1390 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    13/16

    61. Hernan R, Fasheh R, Calabrese C, Frank AJ, Maclean KH et al

    (2003) ERBB2 up-regulates S100A4 and several other prometa-

    static genes in medulloblastoma. Cancer Res 63:140148

    62. Hinds DA, Stuve LL, Nilsen GB, Halperin E, Eskin E, Ballinger

    DG, Frazer KA, Cox DR (2005) Whole-genome patterns of

    common DNA variation in three human populations. Science

    307:10721079

    63. Hirose Y, Aldape K, Bollen A, James CD, Brat D, Lamborn K,

    Berger M, Feuerstein BG (2001) Chromosomal abnormalities

    subdivide ependymal tumors into clinically relevant groups. AmJ Pathol 158:11371143

    64. Hromas R, Klemsz M (1994) The ETS oncogene family in

    development, proliferation and neoplasia. Int J Hematol 59:

    257265

    65. Hu N, Wang C, Hu Y, Yang HH, Giffen C, Tang ZZ, Han XY,

    Goldstein AM, Emmert-Buck MR, Buetow KH, Taylor PR, Lee

    MP (2005) Genome-wide association study in esophageal cancer

    using GeneChip mapping 10 K array. Cancer Res 65:25422546

    66. Huang H, Mahler-Araujo BM, Sankila A, Chimelli L, Yonekawa

    Y, Kleihues P (2000) APC mutations in sporadic medulloblas-

    tomas. Am J Pathol 156:433437

    67. Huang J, Wei W, Zhang J, Liu G, Bignell GR, Stratton MR,

    Futreal PA, Wooster R, Jones KW, Shapero MH (2004) Whole

    genome DNA copy number changes identified by high density

    oligonucleotide arrays. Hum Genomics 1:287299

    68. Hui AB, Takano H, Lo KW, Kuo WL, Lam CN, Tong CY,

    Chang Q, Gray JW, Ng HK (2005) Identification of a novel

    homozygous deletion region at 6q23.1 in medulloblastomas

    using high-resolution array comparative genomic hybridization

    analysis. Clin Cancer Res 11:47074716

    69. Ichimura K, Ohgaki H, Kleihues P, Collins VP (2004) Molecular

    pathogenesis of astrocytic tumors. J Neurooncol 70:137160

    70. Inazawa J, Inoue J, Imoto I (2004) Comparative genomic

    hybridization (CGH)-arrays pave the way for identification of

    novel cancer-related genes. Cancer Sci 95:559563

    71. Ishii N, Sawamura Y, Tada M, Daub DM, Janzer RC, Meagher-

    Villemure M, de Tribolet N, Van Meir EG (1998) Absence of

    p53 gene mutations in a tumor panel representative of pilocytic

    astrocytoma diversity using a p53 functional assay. Int J Cancer

    76:797800

    72. Ishkanian AS, Malloff CA, Watson SK, DeLeeuw RJ, Chi B,

    Coe BP, Snijders A, Albertson DG, Pinkel D, Marra MA, Ling

    V, MacAulay C, Lam WL (2004) A tiling resolution DNA

    microarray with complete coverage of the human genome. Nat

    Genet 36:299303

    73. Jeon IS, Davis JN, Braun BS et al (1995) A variant Ewings

    sarcoma translocation (7;22) fuses the EWS gene to the ETS

    gene ETV1. Oncogene 10:12291234

    74. Johnson RL et al (1996) Human homolog of patched, a candidate

    gene for the basal cell nevus syndrome. Science 272:16681671

    75. Kampa D, Cheng J, Kapranov P, Yamanaka M, Brubaker S,

    Cawley S, Drenkow J, Piccolboni A, Bekiranov S, Helt G,

    Tammana H, Gingeras TR (2004) Novel RNAs identified from

    an in-depth analysis of the transcriptome of human chromosomes

    21 and 22. Genome Res 14:331342

    76. Kaneko Y, Yoshida K, Handa M et al (1996) Fusion of an ETS-

    family gene, E1AF, to EWS by t(17;22)(q12;q12) chromosome

    translocation in an undifferentiated sarcoma of infancy. Genes

    Chromosomes Cancer 15:115121

    77. Kang DE, Soriano S, Xia X, Eberhart CG, De Strooper B et al

    (2002) Presenilin couples the paired phosphorylation of beta-

    catenin independent of axin: implications for beta-catenin

    activation in tumorigenesis. Cell 110:751762

    78. Kapranov P, Cawley SE, Drenkow J, Bekiranov S, Strausberg

    RL, Fodor SP, Gingeras TR (2002) Large-scale transcriptional

    activity in chromosomes 21 and 22. Science 296:916919

    79. Kennedy GC, Matsuzaki H, Dong S, Liu WM, Huang J, Liu G,

    Su X, Cao M, Chen W, Zhang J, Liu W, Yang G, Di X, Ryder T,

    He Z, Surti U, Phillips MS, Boyce-Jacino MT, Fodor SP, Jones

    KW (2003) Large-scale genotyping of complex DNA. Nat

    Biotechnol 21:12331237

    80. Korshunov A, Savostikova M, Ozerov S (2002) Immunohisto-

    chemical markers for prognosis of average-risk pediatric medul-

    loblastomas. The effect of apoptotic index, TrkC, and c-myc

    expression. J Neurooncol 58:271279

    81. Kovar H, Jug G, Aryee DN et al (1997) Among genes involvedin the RB dependent cell cycle regulatory cascade, the p16 tumor

    suppressor gene is frequently lost in the Ewing family of tumors.

    Oncogene 15:22252232

    82. Kramer DL, Parmiter AH, Rorke LB, Sutton LN, Biegel JA

    (1998) Molecular cytogenetic studies of pediatric ependymomas.

    J Neurooncol 37:2533

    83. Krzywinski M, Bosdet I, Smailus D, Chiu R, Mathewson C,

    Wye N, Barber S, Brown-John M, Chan S, Chand S, Cloutier A,

    Girn N, Lee D, Masson A, Mayo M, Olson T, Pandoh P, Prabhu

    AL, Schoenmakers E, Tsai M, Albertson D, Lam W, Choy CO,

    Osoegawa K, Zhao S, de Jong PJ, Schein J, Jones S, Marra MA

    (2004) A set of BAC clones spanning the human genome.

    Nucleic Acids Res 32:36513660

    84. Leung C, Lingbeek M, Shakhova O, Liu J, Tanger E,

    Saremaslani P et al (2004) Bmi1 is essential for cerebellar

    development and is overexpressed in human medulloblasto-

    mas. Nature 428:337341

    85. Lin PP, Brody RI, Hamelin AC, Bradner JE, Healey JH, Ladanyi

    M (1999) Differential transactivation by alternative EWS-FLI1

    fusion proteins correlates with clinical heterogeneity in Ewings

    sarcoma. Cancer Res 59:14281432

    86. Lindsey JC, Lusher ME, Anderton JA, Bailey S, Gilbertson RJ et

    al (2004) Identification of tumor-specific epigenetic events in

    medulloblastoma development by hypermethylation profiling.

    Carcinogenesis 25:661668

    87. Lipshutz RJ, Fodor SP, Gingeras TR, Lockhart DJ (1999) High

    density synthetic oligonucleotide arrays. Nat Genet 21(1 Suppl):

    2024

    88. Lockhart DJ, Winzeler EA (2000) Genomics, gene expression

    and DNA arrays. Nature 405:827836

    89. Losi L, Polito P, Hagemeijer A, Buonamici L, Van den Berghe

    H, Dal Cin P (1998) Intracranial germ cell tumor (embryonal

    carcinoma with teratoma) with complex karyotype including

    isochromosome 12p. Virchows Arch 433:571574

    90. Louis DN, Rubio MP, Correa KM, Gusella JF, von Deimling A

    (1993) Molecular genetics of pediatric brain stem gliomas.

    Application of PCR techniques to small and archival brain

    tumor specimens. J Neuropathol Exp Neurol 52:507515

    91. Lusher ME, Lindsey JC, Latif F, Pearson AD, Ellison DW,

    Clifford SC (2002) Biallelic epigenetic inactivation of the

    RASSF1A tumor suppressor gene in medulloblastoma develop-

    ment. Cancer Res 62:59065911

    92. MacDonald TJ, Brown KM, LaFleur B, Peterson K, Lawlor C,

    Chen Y, Packer RJ, Cogen P, Stephan DA (2001) Expression

    profiling of medulloblastoma: PDGFRA and the RAS/MAPK

    pathway as therapeutic targets for metastatic disease. Nat Genet

    29:143152

    93. Maitra A, Roberts H, Weinberg AG et al (2001) Aberrant

    expression of tumor suppressor proteins in the Ewing family of

    tumors. Arch Pathol Lab Med 125:12071212

    94. Marigo V, Davey RA, Zuo Y, Cunningham JM, Tabin CJ et al

    (1996) Biochemical evidence that Patched is the Hedgehog

    receptor. Nature 384:176179

    95. Matsuzaki H, Dong S, Loi H, Di X, Liu G, Hubbell E, Law J,

    Berntsen T, Chadha M, Hui H, Yang G, Kennedy GC, Webster

    TA, Cawley S, Walsh PS, Jones KW, Fodor SP, Mei R (2004)

    Childs Nerv Syst (2006) 22:13791394 1391

  • 8/9/2019 Child CNS

    14/16

    Genotyping over 100,000 SNPs on a pair of oligonucleotide

    arrays. Nat Methods 1:109111

    96. Matsuzaki H, Loi H, Dong S, Tsai YY, Fang J, Law J, Di X,

    Liu WM, Yang G, Liu G, Huang J, Kennedy GC, Ryder TB,

    Marcus GA, Walsh PS, Shriver MD, Puck JM, Jones KW, Mei

    R (2004) Parallel genotyping of over 10,000 SNPs using a one-

    primer assay on a high-density oligonucleotide array. Genome

    Res 14:414425

    97. Maurici D, Perez-Atayde A, Grier HE et al (1998) Frequency

    and implications of chromosome 8 and 12 gains in Ewingsarcoma. Cancer Genet Cytogenet 100:106110

    98. May WA, Gishizky ML, Lessnick SL et al (1993) Ewing

    sarcoma 11;22 translocation produces a chimeric transcrip-

    tion factor that requires the DNA-binding domain encoded

    by FLI1 for transformation. Proc Natl Acad Sci USA

    90:57525756

    99. Mendrzyk F, Radlwimmer B, Joos S, Kokocinski F, Benner A,

    Stange DE, Neben K, Fiegler H, Carter NP, Reifenberger G,

    Korshunov A, Lichter P (2005) Genomic and protein expression

    profiling identifies CDK6 as novel independent prognostic

    marker in medulloblastoma. J Clin Oncol 23:88538862

    100. Mischel PS, Cloughesy TF, Nelson SF (2004) DNA-microarray

    analysis of brain cancer: molecular classification for therapy. Nat

    Rev Neurosci 5:782792

    101. Morin PJ (1999) Beta-catenin signaling and cancer. Bioessays

    21:10211030

    102. Mugneret F, Lizard S, Aurias A et al (1988) Chromosomes in

    Ewings sarcoma. II. Nonrandom additional changes, trisomy

    8 and der(16)t(1;16). Cancer Genet Cytogenet 32:239245

    103. Newcomb EW, Alonso M, Sung T, Miller DC (2000) Incidence

    of p14ARF gene deletion in high-grade adult and pediatric

    astrocytomas. Hum Pathol 31:115119

    104. Nijssen PC, Deprez RH, Tijssen CC, Hagemeijer A, Arnoldus

    EP, Teepen JL, Holl R, Niermeyer MF (1994) Familial anaplastic

    ependymoma: evidence of loss of chromosome 22 in tumor cells.

    J Neurol Neurosurg Psychiatry 57:12451248

    105. Ohno T, Ouchida M, Lee L et al (1994) The EWS gene,

    involved in Ewing family of tumors, malignant melanoma of

    soft parts and desmoplastic small round cell tumors, codes for

    an RNA binding protein with novel regulatory domains.

    Oncogene 9:30873097

    106. Ohno T, Rao VN, Reddy ESP (1994) EWS/Fli-1 chimeric

    protein is a transcriptional activator. Cancer Res 53:5859

    5863

    107. Olayioye MA, Neve RM, Lane HA, Hynes NE (2000) The ErbB

    signaling network: receptor heterodimerization in development

    and cancer. EMBO J 19:31593166

    108. Ozaki T, Paulussen M, Poremba C et al (2001) Genetic

    imbalances revealed by comparative genomic hybridization in

    Ewing tumors. Genes Chromosomes Cancer 32:164171

    109. Pan E, Pellarin M, Holmes E, Smirnov I, Misra A, Eberhart CG

    (2005) Isochromosome 17q is a negative prognostic factor in

    poor-risk childhood medulloblastoma patients. Clin Cancer Res

    11:47334740

    110. Park PC, Taylor MD, Mainprize TG, Becker LE, Ho M, Dura

    WT et al (2003) Transcriptional profiling of medulloblastoma in

    children. J Neurosurg 99:534541

    111. Peter M, Couturier J, Pacquement H et al (1997) A new member

    of the ETS family fused to EWS in Ewing tumors. Oncogene

    14:11591164

    112. Petermann R, Mossier BM, Aryee DN et al (1998) Oncogenic

    EWS-Fli1 interacts with hsRPB7, a subunit of human RNA

    polymerase II. Oncogene 17:603610

    113. Pietsch T, Waha A, Koch A et al (1997) Medulloblastomas of the

    desmoplastic variant carry mutations of the human homologue of

    Drosophila patched. Cancer Res 57:20852088

    114. Pinkel D, Albertson DG (2005) Array comparative genomic

    hybridization and its applications in cancer. Nat Genet 37(Suppl):

    S11S17

    115. Pinkel D, Albertson DG (2005) Comparative genomic hybrid-

    ization. Annu Rev Genomics Hum Genet 6:331354

    116. Platten M, Giordano MJ, Dirven CM, Gutmann DH, Louis DN

    (1996) Up-regulation of specific NF-1 gene transcripts in

    sporadic pilocytic astrocytomas. Am J Pathol 149:621627

    117. Pollack IF, Hamilton RL, Finkelstein SD, Campbell JW,

    Martinez AJ, Sherwin RN, Bozik ME, Gollin SM (1997) Therelationship between TP53 mutations and overexpression of p53

    and prognosis in malignant gliomas of childhood. Cancer Res

    57:304309

    118. Pollack IF, Finkelstein SD, Burnham J, Holmes EJ, Hamilton

    RL, Yates AJ, Finlay JL, Sposto R (2001) Age and TP53

    mutation frequency in childhood malignant gliomas: results in a

    multi-institutional cohort. Cancer Res 61:74047407

    119. Pomeroy SL, Tamayo P, Gaasenbeek M, Sturla LM, Angelo M,

    McLaughlin ME, Kim JY, Goumnerova LC, Black PM, Lau C,

    Allen JC, Zagzag D, Olson JM, Curran T, Wetmore C, Biegel

    JA, Poggio T, Mukherjee S, Rifkin R, Califano A, Stolovitzky G,

    Louis DN, Mesirov JP, Lander ES, Golub TR (2002) Prediction

    of central nervous system embryonal tumor outcome based on

    gene expression. Nature 415:436442

    120. Prall JA, McGavran L, Greffe BS, Partington MD (1995)

    Intracranial malignant germ cell tumor and the Klinefelter

    syndrome. Case report and review of the literature. Pediatr

    Neurosurg 23:219224

    121. Raffel C, Frederick L, OFallon JR, Atherton-Skaff P, Perry A,

    Jenkins RB, James CD (1999) Analysis of oncogene and tumor

    suppressor gene alterations in pediatric malignant astrocytomas

    reveals reduced survival for patients with PTEN mutations. Clin

    Cancer Res 5:40854090

    122. Rand V, Huang J, Stockwell T, Ferriera S, Buzko O, Levy S,

    Busam D, Li K, Edwards JB, Eberhart C, Murphy KM,

    Tsiamouri A, Beeson K, Simpson AJ, Venter JC, Riggins GJ,

    Strausberg RL (2005) Sequence survey of receptor tyrosine

    kinases reveals mutations in glioblastomas. Proc Natl Acad Sci

    USA 102:1434414349

    123. Rasheed BK, Stenzel TT, McLendon RE, Parsons R, Friedman

    AH, Friedman HS, Bigner DD, Bigner SH (1997) PTEN gene

    mutations are seen in high grade but not in low grade gliomas.

    Cancer Res 57:41874190

    124. Ray A, Ho M, Ma J, Parkes RK, Mainprize TG, Ueda S,

    Mclaughlin J et al (2004) A clinical biological model

    predicting survival in medulloblastoma. Clin Cancer Res

    10:76137620

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

    Boyett J, Kepner JL, Look AT (1999) Chromosome arm 6q

    loss is the most common recurrent autosomal alteration

    detected in primary pediatric ependymoma. Genes Chromo-

    somes Cancer 24:230237

    126. Reifenberger J, Wolter M, Weber RG et al (1998) Missense

    mutation in SMOH in sporadic basal cell carcinoma of the skin

    and primitive neuroectodermal tumors of the central nervous

    system. Cancer Res 58:17981803

    127. Rhodes DR, Chinnaiyan AM (2005) Integrative analysis of the

    cancer transcriptome. Nat Genet 37(Suppl):S31S37

    128. Rickert CH, Simon R, Bergmann M, Dockhorn-Dworniczak

    B, Paulus W (2000) Comparative genomic hybridization in

    pineal germ cell tumors. J Neuropathol Exp Neurol 59:815

    821

    129. Rickert CH, Strater R, Kaatsch P, Wassmann H, Jurgens H,

    Dockhorn-Dworniczak B, Paulus W (2001) Pediatric high grade

    astrocytomas show chromosomal imbalances distinct from adult

    cases. Am J Pathol 158:15251532

    1392 Childs Nerv Syst (2006) 22:13791394

  • 8/9/2019 Child CNS

    15/16

    130. Roberts CW, Galusha SA, McMenamin ME et al (2000)

    Haploinsufficiency of Snf5 (integrase interactor 1) predisposes

    to malignant rhabdoid tumors in mice. Proc Natl Acad Sci USA

    97:1379613800

    131. Rogatto SR, Casartelli C, Rainho CA, Barbieri-Neto J (1993)

    Chromosomes in the genesis and progression of ependymomas.

    Cancer Genet Cytogenet 69:146152

    132. Rood BR, Zhang H, Weitman DM, Cogen PH (2002) Hyper-

    methylation of HIC-1 and 17p allelic loss in medulloblastoma.

    Cancer Res 62:37943797133. Rorke LB, Packer R, Biegel J (1995) Central nervous system

    atypical teratoid/rhabdoid tumors of infancy and childhood.

    J Neurooncol 24:2128

    134. Rorke LB, Packer RJ, Biegel JA (1996) Central nervous system

    atypical teratoid/rhabdoid tumors of infancy and childhood:

    definition of an entity. J Neurosurg 85:5665

    135. Rossi MR, Conroy J, McQuaid D, Nowak NJ, Rutka JT, Cowell

    JK (2006) Array CGH analysis of pediatric medulloblastomas.

    Genes Chromosomes Cancer 45:290303

    136. Rutka JT, Taylor M, Mainprize T, Langlois A, Ivanchuk S,

    Mondal S, Dirks P (2000) Molecular biology in the third

    millennium. Neurosurgery 46:10341051

    137. Sachidanandam R, Weissman D, Schmidt SC, Kakol JM, Stein

    LD, Marth G, Sherry S, Mullikin JC, Mortimore BJ, Willey DL,

    Hunt SE, Cole CG, Coggill PC, Rice CM, Ning Z, Rogers J,

    Bentley DR, Kwok PY, Mardis ER, Yeh RT, Schultz B, Cook L,

    Davenport R, Dante M, Fulton L, Hillier L, Waterston RH,

    McPherson JD, Gilman B, Schaffner S, Van Etten WJ, Reich D,

    Higgins J, Daly MJ, Blumenstiel B, Baldwin J, Stange-Thomann

    N, Zody MC, Linton L, Lander ES, Altshuler D, International

    SNP Map Working Group (2001) A map of human genome

    sequence variation containing 1.42 million single nucleotide

    polymorphisms. Nature 409:928933

    138. Sanoudou D, Tingby O, Ferguson-Smith MA, Collins VP,

    Coleman N (2000) Analysis of pilocytic astrocytoma by compar-

    ative genomic hybridization. Br J Cancer 82:12181222

    139. Satge D, Sommelet D, Geneix A, Nishi M, Malet P, Vekemans

    M (1998) A tumor profile in Down syndrome. Am J Med Genet

    78:207216

    140. Schaid DJ, Guenther JC, Christensen GB, Hebbring S, Rosenow

    C, Hilker CA, McDonnell SK, Cunningham JM, Slager SL,

    Blute ML, Thibodeau SN (2004) Comparison of microsatellites

    versus single-nucleotide polymorphisms in a genome linkage

    screen for prostate cancer susceptibility loci. Am J Hum Genet

    75:948965

    141. Schulze A, Downward J (2001) Navigating gene expression using

    microarraysa technology review. Nat Cell Biol 3:E190E195

    142. Segal E, Friedman N, Kaminski N, Regev A, Koller D (2005)

    From signatures to models: understanding cancer using micro-

    arrays. Nat Genet 37(Suppl):S38S45

    143. Sharrocks AD, Brown AL, Ling Y et al (1997) The ETS-domain

    transcription factor family. Int J Biochem Cell Biol 29:13711387

    144. Sorensen PH, Lessnick SL, Lopez-Terrada D et al (1994) A

    second Ewings sarcoma translocation, t(21;22), fuses the EWS

    gene to another ETS-family transcription factor, ERG. Nat Genet

    6:146151

    145. Stephens P, Hunter C, Bignell G, Edkins S, Davies H, Teague J,

    Stevens C, OMeara S, Smith R, Parker A, Barthorpe A, Blow

    M, Brackenbury L, Butler A, Clarke O, Cole J, Dicks E, Dike A,

    Drozd A, Edwards K, Forbes S, Foster R, Gray K, Greenman C,

    Halliday K, Hills K, Kosmidou V, Lugg R, Menzies A, Perry J,

    Petty R, Raine K, Ratford L, Shepherd R, Small A, Stephens Y,

    Tofts C, Varian J, West S, Widaa S, Yates A, Brasseur F, Cooper

    CS, Flanagan AM, Knowles M, Leung SY, Louis DN, Looijenga

    LH, Malkowicz B, Pierotti MA, Teh B, Chenevix-Trench G,

    Weber BL, Yuen ST, Harris G, Goldstraw P, Nicholson AG,

    Futreal PA, Wooster R, Stratton MR (2004) Lung cancer:

    intragenic ERBB2 kinase mutations in tumors. Nature

    431:525526

    146. Stephens P, Edkins S, Davies H, Greenman C, Cox C, Hunter C,

    Bignell G, Teague J, Smith R, Stevens C, OMeara S, Parker A,

    Tarpey P, Avis T, Barthorpe A, Brackenbury L, Buck G, Butler

    A, Clements J, Cole J, Dicks E, Edwards K, Forbes S, Gorton M,

    Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jones D,

    Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R,

    Raine K, Shepherd R, Small A, Solomon H, Stephens Y, Tofts C,Varian J, Webb A, West S, Widaa S, Yates A, Brasseur F, Cooper

    CS, Flanagan AM, Green A, Knowles M, Leung SY, Looijenga

    LH, Malkowicz B, Pierotti MA, Teh B, Yuen ST, Nicholson AG,

    Lakhani S, Easton DF, Weber BL, Stratton MR, Futreal PA,

    Wooster R (2005) A screen of the complete protein kinase gene

    family identifies diverse patterns of somatic mutations in human

    breast cancer. Nat Genet 37:590592

    147. Suarez-Merino B, Hubank M, Revesz T, Harkness W, Hayward

    R, Thompson D, Darling JL, Thomas DG, Warr TJ (2005)

    Microarray analysis of pediatric ependymoma identifies a cluster

    of 112 candidate genes including four transcripts at 22q12.1

    q13.3. Neuro-oncol 7:2031

    148. Sung T, Miller DC, Hayes RL, Alonso M, Yee H, Newcomb EW

    (2000) Preferential inactivation of the p53 tumor suppressor

    pathway and lack of EGFR amplification distinguish de novo

    high grade pediatric astrocytomas from de novo adult astrocy-

    tomas. Brain Pathol 10:249259

    149. Sure U, Ruedi D, Tachibana O, Yonekawa Y, Ohgaki H,

    Kleihues P, Hegi ME (1997) Determination of p53 mutations,

    EGFR overexpression, and loss of p16 expression in pediatric

    glioblastomas. J Neuropathol Exp Neurol 56:782789

    150. Taipale J, Beachy PA (2001) The Hedgehog and Wnt signalling

    pathways in cancer. Nature 411:349354

    151. Taipale J, Chen JK, Cooper MK, Wang B, Mann RK,

    Milenkovic L et al (2000) Effects of oncogenic mutations in

    Smoothened and Patched can be reversed by cyclopamine.

    Nature 406:10051009

    152. Tamber MS, Rutka JT (2003) Pediatric supratentorial high grade

    gliomas. Neurosurg Focus 14:e1 (serial online)

    153. Tanaka K, Iwakuma T, Harimaya K et al (1997) EWS-Fli1

    antisense oligodeoxynucleotide inhibits proliferation of human

    Ewings sarcoma and primitive neuroectodermal tumor cells.

    J Clin Invest 99:239247

    154. Tarkkanen M, Kiuru-Kuhlefelt S, Blomqvist C et al (1999)

    Clinical correlations of genetic changes by comparative genomic

    hybridization in Ewing sarcoma and related tumors. Cancer

    Genet Cytogenet 114:3541

    155. Taylor MD, Gokgoz N, Andrulis IL et al (2000) Familial

    posterior fossa brain tumors of infancy secondary to germ-line

    mutation of the hSNF5 gene. Am J Hum Genet 66:14031406

    156. Taylor MD, Mainprize TG, Rutka JT (2000) Molecular insight

    into medulloblastoma and central nervous system primitive

    neuroectodermal tumor biology from hereditary syndromes: a

    review. Neurosurgery 47:888901

    157. Taylor MD, Mainprize TG, Squire JA, Rutka JT (2001)

    Molecular genetics of pineal region neoplasms. J Neurooncol

    54:219238

    158. Taylor MD, Liu L, Raffel C, Hui CC, Mainprize TG, Zhang X,

    Rutka JT (2002) Mutation in SUFU predispose to medulloblas-

    toma. Nat Genet 31:306310

    159. Taylor MD, Mainprize TG, Rutka JT (2003) Bio