Embryonal Cns

Embed Size (px)

Citation preview

  • 8/13/2019 Embryonal Cns

    1/13

    Advances in the Diagnosis, Molecular Genetics,and Treatment of Pediatric Embryonal CNS Tumors

    TOBEY J. MACDONALD,a BRIAN R. ROOD,a MARIA R. SANTI,b GILBERT VEZINA,c

    KIMBERLY BINGAMAN,d PHILIP H. COGEN,d ROGER J. PACKERe

    Departments of aHematology/Oncology, bPathology, cRadiology, dNeurosurgery, and eNeurology,

    Childrens Hospital National Medical Center, Washington, DC, USA

    Key Words. Primitive neuroectodermal tumor Medulloblastoma Atypical teratoid/rhabdoid tumor Diagnosis Molecular genetics Treatment

    ABSTRACT

    Embryonal central nervous system (CNS) tumors

    are the most common group of malignant brain tumors

    in children. The diagnosis and classification of tumors

    belonging to this family have been controversial; how-

    ever, utilization of molecular genetics is helping to

    refine traditional histopathologic and clinical classifica-

    tion schemes. Currently, this group of tumors includes

    medulloblastomas, supratentorial primitive neuroecto-

    dermal tumors, atypical teratoid/rhabdoid tumors,

    ependymoblastomas, and medulloepitheliomas. While

    the survival of older children with nonmetastatic

    medulloblastomas has improved considerably within

    the past two decades, the outcomes for infants and for

    those with metastatic medulloblastomas or other high-

    risk embryonal CNS tumors remain poor. It is antici-

    pated that the emerging field of molecular biology will

    greatly aid in the future stratification and therapy for

    pediatric patients with malignant embryonal tumors. In

    this review, recent advances in the diagnosis, molecular

    genetics, and treatment of the most common pediatric

    embryonal CNS tumors are discussed. The Oncologist

    2003;8:174-186

    The Oncologist 2003;8:174-186 www.TheOncologist.com

    Correspondence: Tobey J. MacDonald, M.D., Childrens Hospital National Medical Center, Department ofHematology/Oncology, 111 Michigan Avenue, NW, Washington, DC 20010, USA. Telephone: 202-884-2800; Fax: 202-884-5685; e-mail: [email protected] Received October 21, 2002; accepted for publication January 14, 2003. AlphaMedPress 1083-7159/2003/$12.00/0

    INTRODUCTIONEmbryonal central nervous system (CNS) tumors com-

    prise the most common group of childhood malignant brain

    tumors (21%) [1]. The World Health Organization (WHO)

    classification of tumors recognizes the following entities

    within this group: medulloblastoma (MB), supratentorial

    primitive neuroectodermal tumor (PNET), atypical teratoid/rhabdoid tumor (AT/RT), ependymoblastoma, and medul-

    loepithelioma [2]. MBs, PNETs, and ependymoblastomas

    share a histologically similar, undifferentiated morphology,

    while medulloepitheliomas and AT/RTs have distinctly dif-

    ferent histologies and appear to evolve by different genetic

    TheOncologist

    LEARNING OBJECTIVES

    After completing this course, the reader will be able to:

    1. Recognize the classification, clinical presentation, and diagnosis of embryonal CNS tumors.

    2. Explain the important molecular genetic alterations identified in embryonal CNS tumors.

    3. Describe the current management and novel treatment strategies for embryonal CNS tumors.

    Access and take the CME test online and receive one hour of AMA PRA category 1 credit at CME.TheOncologist.comCMECME

    This material is protected by U.S. Copyright law.

    Unauthorized reproduction is prohibited.

    For reprints contact: [email protected]

  • 8/13/2019 Embryonal Cns

    2/13

    pathways. The incidence of CNS embryonal tumors is con-

    stant from infancy to 3 years of age (11.6 to 10.2 per mil-

    lion) and then steadily declines thereafter [1]. MBs, PNETs,

    and AT/RTs make up the majority of these tumors, the

    remaining being rare, and thus are the focus of this review.

    Controversy exists regarding the division between MBs and

    PNETs, but emerging molecular, biologic, and clinical evi-

    dence supports the separation of these tumors [3]. The inci-

    dence and classification of the more recently described

    entity, AT/RT, is also evolving due in large part to the

    expanded use of diagnostic molecular genetics. Historically,

    AT/RTs have been confused with MBs or PNETs.

    Treatment of these tumors has traditionally relied on

    surgery and radiation therapy (RT). More recently, chemother-

    apy has been utilized to improve outcome and/or delay or

    reduce the dose of RT in an attempt to lessen its neurotoxic

    effects. While the survival of older children with nonmetasta-

    tic MBs has improved considerably within the past two

    decades, the outcomes for infants and for those with metasta-

    tic MBs or other high-risk embryonal CNS tumors remain

    poor. It is hoped that the field of molecular biology will aid in

    the development of novel therapeutics that target specific char-

    acteristics of individual tumors, while minimizing toxicity to

    normal organ systems. This review discusses important

    advances in the diagnosis, molecular genetics, and treatment of

    the most common pediatric embryonal CNS tumors.

    MEDULLOBLASTOMAS AND PRIMITIVE

    NEUROECTODERMAL TUMORSMedulloblastomas account for 40% of all posterior

    fossa tumors and 15%-20% of all childhood brain tumors.

    The peak incidence occurs between 3 and 4 years of age,

    with a male predilection of 1.5- to two-fold [1]. PNETs con-

    stitute 2% of all childhood brain tumors and are most often

    located in the cerebrum, suprasellar, or pineal region of chil-

    dren in their first decade of life [2]. Metastatic disease at

    diagnosis occurs in 11%-43% of MB/PNET cases and is one

    of the most important clinical predictors of outcome [4].

    Extraneural spread of MBs/PNETs is an uncommon event,

    with bone, bone marrow, lymph nodes, liver, and lung

    involvement occurring in decreasing order of frequency.

    Clinical Presentation

    Medulloblastomas arise from the cerebellum, typically

    growing into the fourth ventricle. Patients often present with

    hydrocephaly and raised intracranial pressure (ICP) symp-

    toms, such as headache, lethargy, and morning vomiting.

    Infants in whom the cranial sutures have not fused can pre-

    sent with increasing head circumferences. Cerebellar inva-

    sion results in ataxia and dysmetria. Patients with PNETs

    present with symptoms dependent upon tumor location.

    Paresis and seizures can occur with tumors of the cerebral

    cortex, raised ICP symptoms occur with tumors that obstruct

    cerebrospinal fluid (CSF) flow, and endocrinopathies or

    visual deficits may result from suprasellar tumors.

    Neuropathologic Diagnosis

    The classic histologic appearance of an MB is that of

    densely packed cells with hyperchromatic nuclei, indis-

    cernible cytoplasms, and numerous mitoses (Fig. 1A). Homer

    Wright rosettes and neuroblastic differentiation are observed

    in a minority of cases (Fig. 1B). These tumors may be

    strongly immunoreactive for vimentin and at least focally for

    synaptophysin [5]. Although classic MB is the most common

    form, the WHO classification of CNS tumors describes three

    additional subtypes of MB [2] (Table 1). These subtypes

    include large cell, occurring in approximately 4% of cases,

    desmoplastic (Fig. 1C and D), and the rare MB variant char-

    acterized by extensive nodularity and advanced neuronal dif-

    ferentiation, also known as cerebellar neuroblastoma [2, 6].

    PNETs are histologically similar to classic MBs [2, 7].

    Nuclear polymorphism, brisk mitotic activity, and necrosis

    may be present. Rarely, Homer Wright or Flexner-

    Wintersteiner rosettes are seen. Fields of neuronal cells, glial

    cells, ependymal canals, and striated muscle or melanin-

    bearing cells may be identified, confirming divergent differ-

    entiation along neuronal, astrocytic, ependymal, muscular,

    or melanocytic lines, respectively [8].

    Molecular Genetics and NeurobiologyMolecular biology has augmented traditional histo-

    pathologic and clinical classification schemes by providing

    further insight into the biological diversity of MBs/PNETs.

    This emerging field is expected to have a great impact on

    the diagnosis, classification, and prognosis of MBs/PNETs

    as well as aid in the rational development of innovative

    molecularly targeted therapies. A summary of the most com-

    mon molecular genetic alterations recognized in MBs/PNETs

    is shown in Table 2.

    Expression of the neurotrophin-3 receptor trkCwas the

    first molecular alteration in MBs to be correlated with out-

    come [9]. Neurotrophin receptors regulate cell differentia-

    tion, growth, and apoptosis in the developing cerebellum.

    TrkC activation in MB cells induces apoptosis by initiating

    c-jun and c-fos early gene expression [10]. trkCexpression

    has been found in up to 48% of MB cases [9, 11]. High trkC

    expression is the single most powerful independent predic-

    tor of favorable outcome, with 5-year survival rates as high

    as 89%, compared with 46% for those patients with low

    trkCexpression levels [9, 11].

    High expression of the erbB-2 (c-erbB-2) oncogene

    product, HER2, a member of the epidermal growth factor

    175 Pediatric Embryonal CNS Tumors

  • 8/13/2019 Embryonal Cns

    3/13

    receptor family, correlates with

    poor outcome in MB patients.

    HER2 expression has been found

    in 84% of MB cases, and in those

    patients with more than 50% posi-tive tumor cells, the 10-year sur-

    vival rate was 10%, compared with

    48% for all others [12]. Low

    expression level of the MYCC

    (C-myc) oncogene is predictive of

    greater survival in MB patients

    [13]. MYCC expression has been

    detected in 42% of MB cases. A

    recent study showed that MYCC

    amplification occurs in only 5% of

    MB cases; however, all patientswith this amplification died of

    aggressive disease within 7 months

    of diagnosis [14].

    The nevoid basal cell carci-

    noma syndrome (NBCCS, Gorlins

    syndrome) is an autosomal domi-

    nant disease resulting from mutations of the PTCHgene on

    chromosome 9q22.3. This mutation leads to the develop-

    ment of MB in about 4% of affected patients. Similarly,

    NBCCS is responsible for 1%-2% of all MBs. Studies have

    shown PTCH mutations in about 10% of sporadic MB

    cases, particularly in desmoplastic MBs [15]. PTCH

    MacDonald, Rood, Santi et al. 176

    Figure 1. Histologic features of medul-

    loblastomas. Undifferentiated, classic

    medulloblastoma (A) is characterized

    by patternless sheets of small round

    hyperchromatic cells. Homer Wright

    rosettes (B), the histologic expression

    of neuroblastic differentiation, are

    seen in a minority of cases. In desmo-plastic lesions, the tumor cells are

    compressed into slender columns (C)

    or are organized in nodular zones

    (arrow) (D). (Hematoxylin-eosin stain,

    200magnification).

    Table 1. Histopathologic subtypes of medulloblastomas by WHO classification of CNS tumors

    Medulloblastoma subtype Histologic characteristics IHC+

    Classic [2, 5] High cell density, numerous mitoses, hyperchromatic nuclei, scant cytoplasm Vm, Sn

    Extensive nodularity and Nodules with uniform cells resembling neurocytes of neurocytoma; rare variant NSE, Sn, Nfneuronal differentiation [2, 6]

    Desmoplastic [2] Reticulin-free nodules (pale islands) with uniform cells of low mitotic rate, NSE, Sn, Nfsurrounded by reticulin and mitotically active, hyperchromatic irregular cells

    Large cell [2] Sheets and lobules of round cells with pleiomorphic nuclei, prominent nucleoli, Vm, Snabundant cytoplasm, high mitoses, apoptosis and necrosis; background anaplasiamay be observed.

    Abbreviations: IHC+ = positive immunoreactivity; Vm = vimentin; Sn = synaptophysin; NSE = neuron-specific enolase; Nf = neurofilaments.

  • 8/13/2019 Embryonal Cns

    4/13

    encodes a membrane receptor important for cell growth in

    the developing cerebellum. Experimental models have

    shown that loss of p53 accelerates the development of MBs

    in mice heterozygous for PTCH[16], indicating that PTCH

    acts as a tumor suppressor gene. Sonic hedgehog (SHH),

    a major ligand for the PTCH receptor, is considered a

    putative oncogene.

    Loss of genetic material from the short arm of chromo-some 17 (17p) is the most common cytogenetic abnormality

    in MBs, occurring in 35%-50% of cases [17]. Among the

    genes localized to the common breakpoint at 17p13.3,HIC-

    1 is the leading tumor suppressor gene candidate inactivated

    by 17p deletion. HIC-1 encodes for a zinc finger transcrip-

    tional repressor whose expression is upregulated by p53 and

    is silenced by hypermethylation. Hypermethylation of the

    HIC-1 gene is a frequent event in MB that predicts for a poor

    outcome [18]. Other frequent cytogenetic abnormalities

    include deletions of regions on chromosomes 10q and 11 as

    well as rearrangements of chromosomes 3, 14, 10, 6, 13, 18,

    and 22 [19, 20].

    Despite similar histological appearances, many of the

    molecular genetic aberrations found in MBs are absent in

    PNETs. For example, loss of genetic material from chromo-

    some 17p is not found in PNETs [21]. Patterns of aberrant

    methylation in the region of the 17p breakpoint cluster of

    MBs are also absent [22]. Recent microarray studies have

    revealed that MBs and PNETs could be separated based on

    their specific patterns of gene expression [3]. Furthermore,

    this work illustrated that the sporadic form of desmoplastic

    MB is molecularly similar to that of MB associated with

    NBCCS, yet distinct from classic MB, predominantly due todifferential expression of the PTCH/SHH genes. Most

    importantly, the clinical outcomes of children with MBs

    were best predicted by the gene expression profile of the

    individuals tumor.

    Using similar methodology, another study compared

    gene expression profiles of metastatic (M+) and non-

    metastatic (M0) MBs. This analysis discovered that the

    platelet-derived growth factor receptor alpha (PDGFR-)

    and theRas/mitogen-activated protein (MAP) kinase path-

    way genes were significantly upregulated in M+ tumors

    [23]. This finding suggests that the PDGFR- and

    Ras/MAP kinase signal transduction pathways may be

    rational therapeutic targets for M+ disease.

    Neuroradiographic Findings

    The imaging features of MBs/PNETs are fairly homoge-

    neous throughout the CNS. On T1-weighted images, the solid

    components generally have low signals and strong contrastenhancements. On T2-weighted images, the solid component

    signals are intermediate between gray and white matter; on

    fast fluid-attenuated inversion recovery (FLAIR) images, the

    signals are isointense to gray matter. In contrast, most other

    CNS tumors tend to have T2-weighted and FLAIR signal that

    are greater than gray matter [24]. MBs typically arise in the

    cerebellar vermis and roof of the fourth ventricle, growing for-

    ward into the fourth ventricle, which is displaced anteriorly

    (Fig. 2A and 2B). Invasion of the dorsal brain stem or exten-

    sion into the medial cerebellar hemisphere may occur. MBs

    are typically 3-5 cm in maximal diameter. In older children

    and adolescents, MBs have a tendency to present either in the

    lateral cerebellar hemisphere or near the cerebellopontine

    angle cistern [24]. Atypical imaging features include an exten-

    sive or complete lack of enhancement in up to 25% of lesions,

    cystic or large necrotic areas, and hemorrhage. On computer-

    ized tomography (CT) scans, MBs have a hyperdense appear-

    ance compared with the cerebellum, and calcifications are

    seen in approximately 10% of cases [25].

    PNETs replicate the appearance of MBs (Fig. 3A-3C).

    However, these lesions are generally larger and more com-

    monly display large cystic/necrotic areas. They are typically

    well defined rather than infiltrative, most often located in thefrontoparietal region, and can arise either cortically or in the

    deep periventricular white matter. Calcifications and hemor-

    rhage are more common, especially within the larger cystic

    or necrotic foci. These characteristics result in more hetero-

    geneous magnetic resonance imaging (MRI) features,

    including areas of high T1-weighted signal (hemorrhage)

    and a mixed low and high T2-weighted signal (high cellu-

    larity and cystic, necrotic, and/or hemorrhagic changes)

    [26]. Peritumoral edema is common, though often minimal

    given the large size of these tumors [27].

    177 Pediatric Embryonal CNS Tumors

    Table 2. Common molecular alterations detected in MB and PNET

    Molecular alteration Detected rate Clinical association

    trkC[2, 3, 9-11] 48% of MB cases Low expression unfavorable outcome

    erbB-2 (HER2) [12] 84% of MB cases High expression unfavorable outcome

    MYCC[13, 14] 42% of MB/PNET cases High expression unfavorable outcome

    PTCH[2, 3, 15, 16] 8%-10% of MB cases Mutation development of sporadic and nonsporadic desmoplastic MB

    17p [2, 17-22] 35%-50% of MB cases Deletion unknown significance; putative tumor suppressor gene locus

  • 8/13/2019 Embryonal Cns

    5/13

  • 8/13/2019 Embryonal Cns

    6/13

    nodules or carpet-like coverings of the meningeal sur-

    faces of the brain and spinal cord. However, nonenhanc-

    ing metastatic disease can also be present, especially when

    the primary tumor does not enhance. The nonenhancing

    deposits are often only identified on T2-weighted images

    as areas of distortion of the subarachnoid spaces and can

    also be seen as areas of abnormal signal on FLAIR or

    diffusion images.

    Diffusion-weighted imaging, which reflects Brownian

    diffusion of water molecules, reveals abnormal restriction

    of water movement in most MBs/PNETs. In contrast to

    most CNS tumors, MBs/PNETs are hyperintense on diffu-

    sion-weighted images. The restricted diffusion character-

    istics likely reflect the high cellularity and dense packing

    of MBs/PNETs [26]. The MR spectroscopy (MRS) signa-

    tures of MBs/PNETs reflect that of malignant tumors and

    are not as specific as the imaging features on conventional

    and diffusion images. In general, choline levels are

    markedly increased, N-acetyl aspartate (NAA) is either

    markedly decreased or absent, and lactate/lipid moieties

    can be identified. Choline is a cellular membrane marker;

    its increase reflects increased membrane turnover within

    the tumor. NAA is a neuronal marker; its diminution or

    absence confirms the lack of neuronal differentiation of

    MBs/PNETs. Lactate is a product of anaerobic glycolysis

    and indicates the presence of necrosis or nonaerobic

    cellular metabolism.

    Therapeutic Considerations

    Clinical Prognostic Factors

    Treatment groups for MB are designated high risk and

    average risk based upon the criteria of age greater than or

    less than 3 years, residual tumor greater than or less than

    1.5 cm2, and the presence or absence of metastatic disease

    on neuroimaging or CSF sampling [4, 24]. Age younger

    than 3 years is predictive of poor outcome. One explanation

    for this is that younger children more commonly present

    with metastatic disease [28]; however, they are also less

    likely to be treated with conventional doses of RT [29] and

    are more likely to have subtotal tumor resection [30].

    Extent of resection correlated with better survival for

    patients without metastatic disease in the Childrens Cancer

    Group (CCG) study 921 [31]. Metastatic disease at diagno-

    sis has been repeatedly correlated with poor survival, the

    exception being M1 disease, defined as only CSF cytology

    positive for MB cells [4, 32]. PNETs are considered high

    risk regardless of the patients age, extent of resection, or

    the presence or absence of metastatic disease at diagnosis,

    and as such, are treated in a similar fashion as high-risk

    MBs as outlined below.

    Surgery

    Most MBs are located in the midline of the fourth ven-

    tricle and/or cerebellar vermis, with associated important

    hydrocephalus. If the child presents in extremis from his or

    her hydrocephalus, an emergency ventriculostomy should

    be performed through a frontal burr hole, often at the bed-

    side using conscious sedation. The CSF may then be sam-

    pled for tumor cells as well as drained to a level sufficient to

    relieve the acute symptoms. If the child is not obtunded and

    responds to intravenous corticosteriods alone, a burr hole

    can be placed in the occipital skull at the time of the tumor

    resection and an external ventricular drain placed [33]. In

    the rare instances where hydrocephalus is not initially pre-

    sent, a burr hole should usually be placed anyway at the time

    of tumor resection to allow bedside ventriculostomy should

    postoperative swelling result in CSF obstruction. The child

    is usually operated upon in the prone position: we favor the

    use of a craniectomy rather than replacing the bone flap, for

    these highly malignant tumors often produce considerable

    posterior fossa edema postoperatively. It may be necessary

    to remove the posterior arch of the first cervical vertebra to

    gain access below the cerebellar tonsils.

    Using the operating microscope, the cerebellar tonsils

    should be carefully separated following the dural opening,

    and the floor of the fourth ventricle can be identified and

    protected with a cottonoid pledgett. The majority of these

    tumors arise from this region, and their attachment may be

    identified. The bulk of the tumor can then be resected by

    splitting the vermis and retracting the cerebellar hemi-spheres. Useful surgical adjuncts include the Cavitron ultra-

    sonic aspirator. Care must be taken to avoid undue

    dissection of the roof of the third ventricle, which results in

    ocular pareses, but the tumor must be fully resected from

    this location to remove the inferior third ventricular obstruc-

    tion that is almost always present. Dissection at the junction

    of the cerebellar peduncles and brainstem may be the origin

    of the phenomenon of postoperative mutism [34]. In gen-

    eral, an attempt should be made to remove the entire tumor

    [35]. This may not be possible when there is encasement of

    the posterior inferior cerebellar artery or extensive involve-

    ment of the brainstem. However, it is sometimes possible

    that residual tumor detected on the postoperative MRI scan

    can be safely resected, and under these circumstances, a sec-

    ond operation should be attempted to achieve a complete

    resection in patients with nonmetastatic disease. If there is

    already leptomeningeal dissemination seen at the time of the

    resection, then no attempt should be made to route out every

    last cell of the primary mass. Common postoperative

    deficits in addition to mutism include ataxia, hemiparesis,

    and sixth nerve palsy, which generally resolve over time

    [36]. Approximately 60%-75% of children in whom a total

    179 Pediatric Embryonal CNS Tumors

  • 8/13/2019 Embryonal Cns

    7/13

    or near-total resection of the mass is achieved will not

    require permanent CSF diversion. The remainder of these

    children should undergo placement of a ventricular shunt

    generally at day 5-7 postoperatively, when the CSF has

    cleared from blood and debris, and it is clear that a

    permanent implant will be required.

    Medulloblastomas that present in the cerebellopontine

    angle, once classified as reticulum cell sarcomas (primarily

    now known as the desmoplastic variant), should be

    approached through a laterally placed incision and craniec-

    tomy. These tumors are generally completely resectable, as

    they do not involve the fourth ventricle, and often present

    with hydrocephalus. This is also a common location for

    AT/RTs, although this latter type tends to envelop the cra-

    nial nerves, arteries, and brainstem, making their resection

    more problematic. Supratentorial PNETs should be

    approached through a craniotomy placed in relation to their

    site of origin. These tumors are most often extremely largeand vascular. An attempt should be made to resect the

    entire primary mass, unless there is widespread lep-

    tomeningeal disease. The use of intraoperative neuronavi-

    gation (frameless stereotactic guidance) can be quite

    helpful in the resection of these tumors.

    Radiation and Chemotherapy

    The cornerstone of MB/PNET treatment has been RT of

    the primary tumor site. However, given the propensity of

    MBs/PNETs to spread, the addition of craniospinal radio-

    therapy (csRT) for prophylactic treatment of metastasis hasbeen necessary to maximize survival [37]. Unfortunately,

    the neurocognitive and endocrine effects resulting from irra-

    diation of the developing neuraxis have presented a high

    price for this protection. In an attempt to lessen RT-induced

    neurotoxicity, clinical trials utilizing adjuvant chemotherapy

    have been explored.

    Medulloblastomas respond to a range of alkylator and

    platinum-based drugs. A CCG study of patients with aver-

    age-risk MBs reduced the csRT dose from the standard

    3,600 cGy to 2,340 cGy (total boost 5,580 cGy) and added

    adjuvant chemotherapy consisting of vincristine, cisplatin,

    and lomustine (CCNU). Progression-free survival was 86%

    at 3 years and 79% at 5 years [38]. These rates compared

    favorably with historical controls. A CCG trial using an

    identical RT dose followed by a randomization between the

    chemotherapy described above and one substituting

    cyclophosphamide for the CCNU was recently completed.

    These data are awaited to confirm the promising results for

    reduced-dose csRT in this group of patients.

    Despite this reduction in csRT, neurocognitive deficits

    were still noted. Patients who underwent longitudinal intel-

    ligence testing demonstrated an estimated rate of change

    from baseline of -4.3 Full Scale Intelligence Quotient points

    per year, -4.2 Verbal IQ points per year, and -4.0 Nonverbal

    IQ points per year (p < 0.001 for all three outcomes).

    Females, children aged less than 7 years, and those with

    higher baseline IQs were at greatest risk [39].

    Doses of 3,600 cGy csRT with total tumor boost to

    5,400 cGy have been used to treat high-risk MBs and

    PNETs in neurodevelopmentally appropriate patients.

    However, when used as the sole postoperative treatment,

    results were dismal. Yet objective responses to chemother-

    apy were observed in up to 50% of patients. Postoperative

    chemoradiotherapy for non-pineal PNETs have produced 5-

    year survivals in approximately one-third of patients, with

    children less than 2 years faring more poorly [30]. Although

    infants with pineal PNETs did poorly, older patients with

    this type of tumor in this location appeared to have a better

    prognosis [30].

    In very young children, for whom the long-term neu-rocognitive sequelae of RT are unacceptable, high-dose

    chemotherapy (HDCT) and autologous stem cell (ACS) sup-

    port have been used in an attempt to delay or obviate the need

    for RT. In a study of 23 relapsed MB patients who received

    HDCT consisting of carboplatin, thiotepa, and etoposide

    with autologous stem cell (ASC) rescue, 3-year event-free

    survival (EFS) and overall survival (OS) rates were 34% and

    46%, respectively [40]. Trials of HDCT and ASC as front-

    line therapy are ongoing in patients less than 3 years of age

    with MBs/PNETs and as therapy following csRT for older

    children with high-risk MBs or PNETs.

    ATYPICAL TERATOID/RHABDOID TUMORS

    Atypical teratoid/rhabdoid tumors, first described by

    Rorke et al. in 1987, are considered by some as a subtype

    of PNET [41-44]. With the wider utilization of immunohis-

    tochemistry and new molecular genetic probes, AT/RTs

    have been increasingly diagnosed, especially in infants and

    very young children [42, 44]. AT/RTs also have been diag-

    nosed in older children and young adults [45-48]. The exact

    incidence of this tumor is unknown, but it has been sug-

    gested that approximately 10%-15% of children less than 3

    years of age thought to have MBs or other forms of PNETs,

    actually had AT/RTs [45-48]. Others have reported that the

    ratio of AT/RTs to other more common PNETs is as low as

    1:4 among children less than 3 years of age [49].

    Clinical Presentation

    AT/RTs present in a similar fashion to other PNETs and

    can arise throughout the nervous system. Approximately

    one-half of patients will have tumors originating in the pos-

    terior fossa, with a possible predilection for the cerebello-

    pontine angle [42, 47]. Supratentorial AT/RTs tend to be

    MacDonald, Rood, Santi et al. 180

  • 8/13/2019 Embryonal Cns

    8/13

    extremely large at the time of diagnosis and may have cys-

    tic/necrotic components [42-48]. The tumors can be intra-

    or extra-axial and often invade adjacent structures. The

    incidence of leptomeningeal dissemination at the time of

    diagnosis has not been firmly established. Early review

    suggested that as much as 30%-40% of patients had lep-

    tomeningeal dissemination, although in more recent studies

    the incidence of dissemination was noted to be closer to

    15% [42-48].

    Neuropathologic Diagnosis

    AT/RTs are malignant embryonal tumors composed of

    rhabdoid cells usually with additional, variable components

    of primitive neuroectodermal, mesenchymal, and epithelial

    cells [42, 44, 50]. The typical rhabdoid cell is medium sized,

    round to oval, with distinct borders, an eccentric nucleus, and

    commonly a prominent nucleolus (Fig. 4). The cytoplasm

    has a fine granular character or may contain a poorly defined

    pink body resembling an inclusion. Variable elements

    from small cells with tapering cytoplasmic tails to large

    bizarre cells may be identified. The primitive neuroectoder-

    mal component may consist of sheets of small round blue

    cells or may display Homer Wright or Flexner-Wintersteiner

    rosettes. The mesenchymal component appears as loose

    arrangements of small spindle cells or tightly arranged in a

    fascicular pattern resembling sarcoma. Epithelial differentia-

    tion is uncommon, and if present, is confined to few gland-

    like spaces. Mitoses are abundant, and field necrosis is

    common. The immunophenotype is broad, as the large rhab-doid cells display a range of immunoreactivity with clusters

    of cells almost always positive for epithelial membrane anti-

    gen and vimentin. Also frequent is reactivity for glial fibril-

    lary acidic protein and cytokeratin, and less frequent is

    reactivity for smooth muscle actin and neurofilament protein.

    The rhabdoid cells are negative for desmin and any of the

    markers for germ cell tumors [42, 44].

    Molecular Genetics and Neurobiology

    Molecular genetic analysis has aided greatly in the diag-

    nosis and understanding of AT/RTs. The vast majority of

    AT/RTs demonstrate monosomy 22 or deletions of chromo-

    some band 22q11 [51, 52]. Other CNS tumors may demon-

    strate chromosome 22 abnormalities, and this abnormality

    alone is not sufficient for diagnosis. MBs and other PNETs

    may show a deletion of chromosome 22, but can be distin-

    guished from AT/RTs by the presence of associated chromo-

    some abnormalities. Eighty-five percent or more of AT/RTs

    show alterations of the hSNF5/INI1 gene [52-54]. The direct

    function of this gene in tumor development is unknown, but

    homozygous inactivation of the hSNF5/INI1 gene likely

    results in altered transcriptional regulation of downstream

    targets by the chromatin remodeling complex (SWI/SNF).

    The mutations in this gene are predominantly point muta-

    tions that result in the coding of a novel stop codon, which

    predicts premature truncation of the protein [53-55].

    181 Pediatric Embryonal CNS Tumors

    Figure 4. Histologic features of an AT/RT. The cells have large

    nuclei with prominent nucleoli (arrowhead), and some cells pos-

    sess abundant eosinophilic cytoplasm (arrow) (A). Vimentin reac-

    tivity (B) is universal, and positive staining for epithelial

    membrane antigen (C) is common in groups of cells (arrow).

    (Hematoxylin-eosin, vimentin, and epithelial membrane antigen

    stains, 400magnification).

  • 8/13/2019 Embryonal Cns

    9/13

    Neuroradiographic Findings

    The CT findings of AT/RTs are relatively characteris-

    tic, but not diagnostic. These tumors are usually hyperdense

    and enhance intensely [42]. Calcifications may occur but

    are not common, while cysts are more common in the

    supratentorial lesions. On MRI imaging (Fig. 5), the T1 sig-

    nal of the solid portion of the tumor is typically isointense;

    there are frequent T1 hyperintense foci (secondary to intra-

    tumoral hemorrhage) and hypointense foci (secondary to

    cystic/necrotic change). AT/RTs commonly display intense

    contrast enhancement. The T2 appearance is heterogeneic.

    The MRS appearance of an AT/RT is similar to that of a

    PNET, with marked elevation of choline and low or absentNAA and creatine; lipids and lactate peaks can often be

    identified.

    Therapeutic Considerations

    To date, the therapy for AT/RTs has been suboptimal.

    Information about response to therapy and outcome has

    been primarily gathered from retrospective reviews of a

    handful of patients [42-48]. An AT/RT registry has added

    some useful information [47]. The role of surgery for

    AT/RTs is unsettled [56]. Although initial reports suggested

    that, because of the age of the patients, the large extent ofthe tumors, and their tendency to be more laterally placed

    in the cerebellopontine angle, total or near-total resection

    was quite uncommon. In the AT/RT registry, six of the

    eight patients who survived for greater than 18 months had

    undergone total resection.

    Given the young age of the patients, chemotherapy has

    been the primary modality of treatment after radiation ther-

    apy [56]. Even after aggressive surgery and chemotherapy,

    overall survival rates for children, especially those less than

    2 years of age, have been extremely poor, with less than 20%

    of patients surviving less than 12 months from diagnosis. A

    variety of different chemotherapeutic agents have been uti-

    lized, but no one agent or combination of agents has been

    shown to be most effective. The majority of children have

    been treated with chemotherapeutic regimens developed for

    infantile brain tumors that have included drugs such as

    cyclophosphamide, cisplatin, etoposide, and vincristine. The

    use of myeloablative doses of chemotherapy, supported

    either by autologous bone marrow transplant or peripheral

    stem cell rescue, has not been shown to increase survival.

    Because of the histological appearance of these tumors,

    another approach has been to utilize sarcoma chemotherapy

    MacDonald, Rood, Santi et al. 182

    Figure 5. Atypical teratoid/rhabdoid tumor in the right cerebello-

    pontine angle. Axial T2-weighted (A), postcontrast axial (B), and

    sagittal (C) T1-weighted images demonstrate a heterogeneous mass

    (A) with central enhancement and dural extension (B, C).

  • 8/13/2019 Embryonal Cns

    10/13

    regimens [56]. In general, these regimens have shown a

    slightly higher overall response rate; however, the majority

    of patients treated with such regimens have been somewhat

    older. In general, the results of chemotherapeutic studies

    suggest that a variety of chemotherapeutic regimens may

    result in tumor stabilization and, for fewer patients, objective

    tumor shrinkages. The benefit of chemotherapy has not been

    durable for most patients. Because of the age of patients,

    radiotherapy has been less widely employed in children with

    AT/RTs [42-48, 56]. Most of the children reported to the

    AT/RT tumor registry that survived for greater than 18

    months received at least local RT [47, 56]. However, conclu-

    sions are difficult to draw, since many of those patients were

    older at the time of diagnosis.

    In summary, therapeutic approaches have been subopti-

    mal, with the majority of patients developing progressive dis-

    ease within 12 months of diagnosis and dying soon after. As

    the prognosis of children with AT/RTs seems to differ from

    those with MBs/PNETs, investigators have suggested that

    AT/RTs be removed from present infant brain tumor proto-

    cols and entered on protocols designed specifically for

    AT/RTs [56]. There is sentiment to use high-dose chemother-

    apy for a shorter period of time and institute at least local

    radiotherapy earlier for patients with localized disease at the

    time of diagnosis. The optimal induction therapy is not clear

    from available data and there is no treatment that has shown

    significant efficacy for children with disseminated disease at

    the time of diagnosis.

    NOVEL THERAPEUTIC STRATEGIES FOR EMBRYONAL

    CNS TUMORS

    The development of therapies with acceptable toxicities

    that can adequately penetrate the CNS yet remain relatively

    unsusceptible to the emergence of tumor resistance is criti-

    cal to improving the outcome of pediatric embryonal CNS

    tumors. Treatment strategies can be broadly separated into

    two categories: methods that increase the total dose of

    drug/radiation delivered to the focal sites of CNS disease

    and novel therapeutics that exploit the specific biological

    characteristics of the tumor. Clinical strategies that are

    currently active are summarized in Table 3.

    High-dose systemic chemotherapy, with ASC or periph-

    eral blood stem cell (PBSC) support, is being evaluated in

    children with CNS tumors. The aim of HDCT is to increase

    the tumors exposure to cytotoxic agents by overcoming the

    limited permeability of the blood-brain barrier (BBB).

    Classic alkylating agents, which generally have nonoverlap-

    ping hematological toxicities, show little cross-resistance,

    and maintain steep and linear dose-response curves, have

    been predominantly investigated by this approach. Because

    of its lipid solubility, thiotepa has been commonly used.

    Initial results with thiotepa and busulfan in 20 children with

    relapsed malignant tumors showed five partial responses

    (4/8 MB/PNET) for an overall response rate of 26% [57].

    A more recent CCG study using carboplatin, thiotepa, and

    etoposide followed by ASC support for 23 patients with

    recurrent MBs reported a 3-year EFS rate of 34% and an OS

    rate of 46% [40]. A subsequent study evaluated this regimen

    in 62 patients with newly diagnosed malignant brain tumors.

    The EFS and OS rates at 3 years were 25% and 40%, respec-

    tively [58]. The most impressive responses were again noted

    in MB/PNET patients. Despite these promising responses,

    the toxicity associated with these regimens has been exces-

    sively high (5%-15% death rate). In an effort to reduce toxi-

    city, more recent investigations have used multiple cycles ofsomewhat lower doses of chemotherapy followed by PBSC

    support. This has decreased transplant-related complications;

    however, the data relating to efficacy from ongoing trials are

    still premature.

    Administration of intrathecal (IT) chemotherapy or coad-

    ministration of systemic chemotherapy with biologic agents

    that disrupt BBB permeability are alternative methods to

    183 Pediatric Embryonal CNS Tumors

    Table 3. Active clinical trials utilizing novel therapeutic strategies for embryonal CNS tumors

    Novel treatment strategy Desired effect Active clinical trials (agent)HDCT and ASC support Penetrate BBB, CNS drug level COG-99702, high-risk patients, closed; COG-99703, infant patients;

    POG-9430, recurrent disease

    IT chemotherapy Prevent or treat LM disease PBTC-001 (mafosfamide); PBTC-005 (busulfan); COG-P9962 (topotecan)

    Radiosensitization RT cytotoxicity COG-99701 (carboplatin/RT)

    BBB disruption CNS drug level COG-09716 (carboplatin/lobradimil)

    Biologic therapy Target essent ial tumor bioactivity PBTC-002 (VEGFR TKI), closed; PBTC-003 (FTI)

    Focal RT RT neurotoxicity PBTC-001 (3-D conformal RT)

    Abbreviations: ASC = autologous stem cell; COG = Childrens Oncology Group; FTI = farnesyl transferase inhibitor; HDCT = high-dose chemotherapy;LM = leptomeningeal; PBTC = Pediatric Brain Tumor Consortium; POG = Pediatric Oncology Group; VEGFR TKI = vascular endothelial growth factorreceptor tyrosine kinase inhibitor.

  • 8/13/2019 Embryonal Cns

    11/13

    increase CNS drug penetration and control leptomeningeal

    disease. The former method had been limited by the lack of

    available active agents that can be given by IT administration.

    The availability of topotecan and mafosfamide, a preactivated

    derivative of cyclophosphamide, has led to renewed interest in

    regional therapy. A European trial with IT mafosfamide (20

    mg) and systemic chemotherapy for disseminated pediatric

    brain tumors demonstrated complete responses in eight of nine

    evaluable patients and, at a median follow-up of 21 months, 11

    of 16 patients remained in complete or partial remission [59].

    For the latter method, bradykinin agonists, such as lobradimil,

    which cause vasodilatation and leakiness of the BBB, have

    been utilized. This agent has been used in conjunction with

    systemic carboplatin for refractory CNS tumors.

    Poorly oxygenated cells comprise a significant portion of

    the total tumor mass and are nearly three times less sensitive

    than well-oxygenated cells to the effects of RT. Investigations

    have thus focused on particles that are less dependent on oxy-gen for their effect, such as neutrons, or agents that enhance

    the effect of radiation-induced free radicals, such as platinum

    agents and halogenated pyrimidines. Topotecan and pacli-

    taxel, members of the camptothecin and taxane classes of

    chemotherapeutic agents, respectively, are under investiga-

    tion for their effects as radiosensitizers. Pediatric trials are

    also investigating gadolinium-texaphyrin, a porphyrin com-

    pound that produces long-lived free radicals, conjugated to

    gadolinium [60]. This conjugate forms a tumor-selective

    radiosensitizer that can be visualized by MRI.

    The delivery and transfer of foreign genes into tumorcells, a process known as gene therapy, has broad implica-

    tions for the treatment of neoplastic diseases. The postmitotic

    environment of the CNS may provide an advantage over other

    tissues in that it allows for the specific uptake of foreign

    genetic material into the genome of the rapidly dividing

    tumor. To date, one study has been completed and reported in

    pediatric CNS tumors. In this phase I study, 12 patients with

    recurrent malignant supratentorial tumors were multiply

    injected in the rim of the resection cavity with murine vector-

    producing cells shedding the retroviral vector containing the

    herpes simplex virus-1 thymidine kinase gene, and then

    treated with cytotoxic ganciclovir [61]. The procedure was

    well tolerated and future trials are planned.

    The advent of STI571 (imatinib mesylate), an inhibitor

    of the bcr-abl fusion protein found in Philadelphia-chromo-

    some-positive leukemias, ushered in a new paradigm for

    cancer treatment based upon the identification of molecular

    targets [62]. Following this model, investigation is under

    way to find molecular targets in MBs/PNETs. A number of

    promising compounds are just entering phase I clinical trials

    in pediatric patients, including tyrosine kinase inhibitors that

    impede growth factor signaling and farnesyl transferase

    inhibitors that block Ras activation.

    It is unclear whether chemotherapy alone can induce

    durable responses in a significant proportion of patients.

    Three-dimensional (3-D) conformal RT is a technique that

    attempts to minimize neurotoxicity by integrating many

    beams, precisely directing RT to the desired site while leav-

    ing untargeted areas minimally exposed. The achievement of

    this goal depends upon precise localization of the tumor and

    normal critical structures by integrating CT or MRI withreproducible positioning of the patient. Intensity-modulated

    radiation therapy (IMRT) is a new conformal technique that

    makes use of 3-D-based treatment planning and nonuniform

    radiation beams. The beams are of greatest intensity within

    the tumor, sparing nearby critical structures. The high-dose

    treatment volume can then be made to conform to an irreg-

    ular target. When compared with conventional RT, IMRT

    delivered 68% of the dose to the auditory apparatus (mean

    dose, 36.7 versus 54.2 Gy), while the overall incidence of

    ototoxicity was lower in the IMRT group [63].

    SUMMARY

    The current treatment of pediatric embryonal CNS

    tumors continues to be very challenging and too frequently

    results in significant long-term sequelae in survivors. This

    is especially true for very young children, the most com-

    mon age group diagnosed with these tumors, in which the

    effects of chemoradiotherapy on the developing neuraxis

    are greatest. Innovative delivery and decreased neurotoxic-

    ity of chemoradiotherapy are major directives for future

    clinical trials. It is also anticipated that the expanded use of

    molecular genetics will help to better stratify patients, tailor

    individual therapy, and aid in the development of targeted

    therapeutics.

    MacDonald, Rood, Santi et al. 184

    REFERENCES

    1 Gurney JG, Bunin GR. CNS and miscellaneous intracranial and

    intraspinal neoplasms. In: Ries LAG, Kosary CL, Hankey BF et

    al., eds. Cancer incidence and survival among children and ado-

    lescents: United States SEER Program 1975-1995. NIH Pub No

    99-4649. Bethesda: National Cancer Institute, 1999:51-63.

    2 Becker LE, Giangaspero F, Rorke LB et al. Embryonal tumours.

    In: Kleihues P, Cavenee WK, eds. World Health Organization

    Classification of Tumours, Pathology and Genetics of Tumours of

    the Nervous System. Lyon, France: IARC Press, 2000:123-148.

    3 Pomeroy SL, Tamayo P, Gaasenbeek M et al. Prediction of

    central nervous system embryonal tumour outcome based on

    gene expression. Nature 2002;415:436-442.

    4 Zeltzer PM, Boyett JM, Finlay JL et al. Metastasis stage,

    adjuvant treatment, and residual tumor are prognostic factors

  • 8/13/2019 Embryonal Cns

    12/13

    for medulloblastoma in children: conclusions from the

    Childrens Cancer Group 921 randomized phase III study.

    J Clin Oncol 1999;17:832-845.

    5 Coffin CM, Braun JT, Wick MR et al. A clinicopathologic

    and immunohistochemical analysis of 53 cases of medul-

    loblastoma with emphasis on synaptophysin expression. Mod

    Pathol 1990;3:164-170.6 Pearl GS, Takei Y. Cerebellar neuroblastoma: nosology as

    it relates to medulloblastoma. Cancer 1981;47:772-779.

    7 Hart MN, Earle KM. Primitive neuroectodermal tumors of

    the brain in children. Cancer 1973;32:890-897.

    8 Gould VE, Jansson DS, Molenaar WM et al. Primitive neu-

    roectodermal tumors of the central nervous system. Patterns

    of expression of neuroendocrine markers, and all classes of

    intermediate filament proteins. Lab Invest 1990;62:498-509.

    9 Segal RA, Goumnerova LC, Kwon YK et al. Expression of

    the neurotrophin receptor TrkC is linked to a favorable out-

    come in medulloblastoma. Proc Natl Acad Sci USA

    1994;91:12867-12871.

    10 Kim JY, Sutton ME, Lu DJ et al. Activation of neurotrophin-

    3 receptor TrkC induces apoptosis in medulloblastomas.

    Cancer Res 1999;59:711-719.

    11 Grotzer MA, Janss AJ, Fung K et al. TrkC expression predicts

    good clinical outcome in primitive neuroectodermal brain

    tumors. J Clin Oncol 2000;18:1027-1035.

    12 Gilbertson RJ, Pearson AD, Perry RH et al. Prognostic sig-

    nificance of the c-erbB-2 oncogene product in childhood

    medulloblastoma. Br J Cancer 1995;71:473-477.

    13 Grotzer MA, Hogarty MD, Janss AJ et al. MYC messenger

    RNA expression predicts survival outcome in childhood

    primitive neuroectodermal tumor/medulloblastoma. ClinCancer Res 2001;7:2425-2433.

    14 Aldosari N, Bigner SH, Burger PC et al.MYCCand MYCN

    oncogene amplification in medulloblastoma. A fluorescence

    in situ hybridization study on paraffin sections from the

    Childrens Oncology Group. Arch Pathol Lab Med

    2002;126:540-544.

    15 Raffel C, Jenkins RB, Frederick L et al. Sporadic medul-

    loblastomas contain PTCH mutations. Cancer Res 1997;57:

    842-845.

    16 Wetmore C, Eberhart DE, Curran T. Loss of p53 but not ARF

    accelerates medulloblastoma in mice heterozygous for

    patched. Cancer Res 2001;61:513-516.

    17 Biegel JA, Janss AJ, Raffel C et al. Prognostic significance of

    chromosome 17p deletions in childhood primitive neuroecto-

    dermal tumors (medulloblastomas) of the central nervous sys-

    tem. Clin Cancer Res 1997;3:473-478.

    18 Rood BR, Zhang H, Weitman DM et al. Hypermethylation of

    HIC-1 and 17p allelic loss in medulloblastoma. Cancer Res

    2002;62:3794-3797.

    19 Biegel JA. Cytogenetics and molecular genetics of childhood

    brain tumors. Neuro-oncol 1999;1:139-151.

    20 Bayani J, Zielenska M, Marrano P et al. Molecular cytogenetic

    analysis of medulloblastomas and supratentorial primitive

    neuroectodermal tumors by using conventional banding, com-

    parative genomic hybridization, and spectral karyotyping.

    J Neurosurg 2000;93:437-448.

    21 Burnett ME, White EC, Sih S et al. Chromosome arm 17p

    deletion analysis reveals molecular genetic heterogeneity in

    supratentorial and infratentorial primitive neuroectodermal

    tumors of the central nervous system. Cancer GenetCytogenet 1997;97:25-31.

    22 Fruhwald MC, ODorisio MS, Dai Z et al. Aberrant hyper-

    methylation of the major breakpoint cluster region in 17p11.2

    in medulloblastomas but not supratentorial PNETs. Genes

    Chromosomes Cancer 2001;30:38-47.

    23 MacDonald TJ, Brown KM, LaFleur B et al. Expression pro-

    filing of medulloblastoma: PDGFRA and the RAS/MAPK

    pathway as therapeutic targets for metastatic disease. Nat

    Genet 2001;29:143-152.

    24 Packer RJ, Cogen P, Vezina G et al. Medulloblastoma: clinical

    and biologic aspects. Neuro-oncol 1999;1:232-250.

    25 Kumar R, Achari G, Banerjee D et al. Uncommon presenta-

    tion of medulloblastoma. Childs Nerv Syst 2001;17:538-542;

    discussion 543.

    26 Erdem E, Zimmerman RA, Haselgrove JC et al. Diffusion-

    weighted imaging and fluid attenuated inversion recovery

    imaging in the evaluation of primitive neuroectodermal

    tumors. Neuroradiology 2001;43:927-933.

    27 Figeroa RE, el Gammal T, Brooks BS et al. MR findings on

    primitive neuroectodermal tumors. J Comput Assist Tomogr

    1989;13:773-778.

    28 Deutsch M. Medulloblastoma: staging and treatment out-

    come. Int J Radiat Oncol Biol Phys 1988;14:1103-1107.

    29 Saran FH, Driever PH, Thilmann C et al. Survival of very

    young children with medulloblastoma (primitive neuroecto-

    dermal tumor of the posterior fossa) treated with craniospinal

    irradiation. Int J Radiat Oncol Biol Phys 1998;42:959-967.

    30 Duffner PK, Horowitz ME, Krischer JP et al. Postoperative

    chemotherapy and delayed radiation in children less than

    three years of age with malignant brain tumors. N Engl J Med

    1993;328:1725-1731.

    31 Albright AL, Wisoff JH, Zeltzer PM et al. Effects of medul-

    loblastoma resections on outcome in children: a report from the

    Childrens Cancer Group. Neurosurgery 1996;38:265-271.

    32 Kortmann RD, Kuhl J, Timmermann B et al. Postoperativeneoadjuvant chemotherapy before radiotherapy as compared

    to immediate radiotherapy followed by maintenance

    chemotherapy in the treatment of medulloblastoma in child-

    hood: results of the German prospective randomized trial HIT

    91. Int J Radiat Oncol Biol Phys 2000;46:269-279.

    33 Lee M, Wisoff JH, Abbott R et al. Management of hydro-

    cephalus in children with medulloblastoma: prognostic fac-

    tors for shunting. Pediatr Neurosurg 1994;2:240-247.

    34 Pollack IF, Polinko P, Albright AL et al. Mutism and pseudo-

    bulbar symptoms after resection of posterior fossa tumors in

    children: incidence and pathophysiology. Neurosurgery

    1995;37:885-893.

    185 Pediatric Embryonal CNS Tumors

  • 8/13/2019 Embryonal Cns

    13/13

    35 Cochrane DD, Gustavsson B, Poskitt KP et al. The surgical

    and natural morbidity of aggressive resection for posterior

    fossa tumors in childhood. Pediatr Neurosurg 1994;20:19-29.

    36 Catsman-Berrevoets CE, Van Donegan HR, Mulder PG et al.

    Tumour type and size are high risk factors for the syndrome

    of cerebellar mutism and subsequent dysarthria. J Neurol

    Neurosurg Psychiatry 1999;67:755-757.

    37 Bouffet E, Bernard JL, Frappaz D et al. M4 protocol for cere-

    bellar medulloblastoma: supratentorial radiotherapy may not

    be avoided. Int J Radiat Oncol Biol Phys 1992;24:79-85.

    38 Packer RJ, Goldwein J, Nicholson HS et al. Treatment of chil-

    dren with medulloblastomas with reduced-dose craniospinal

    radiation therapy and adjuvant chemotherapy: a Childrens

    Cancer Group study. J Clin Oncol 1999;17:2127-2136.

    39 Ris MD, Packer R, Goldwein J et al. Intellectual outcome after

    reduced-dose radiation therapy plus adjuvant chemotherapy for

    medulloblastoma: a Childrens Cancer Group study. J Clin

    Oncol 2001;19:3470-3476.

    40 Dunkel IJ, Boyett JM, Yates A et al. High-dose carboplatin,

    thiotepa, and etoposide with autologous stem-cell rescue for

    patients with recurrent medulloblastoma. Childrens Cancer

    Group. J Clin Oncol 1998;16:222-228.

    41 Lefkowitz IB, Rorke JB, Packer RJ et al. Atypical teratoid

    tumor of infancy: definition of an entity. Ann Neurol

    1987;22:448a-449a.

    42 Rorke LB, Packer RJ, Biegel JA. Central nervous system

    atypical teratoid/rhabdoid tumors of infancy and childhood:

    definition of an entity. J Neurosurg 1996;85:56-65.

    43 Biegel JA, Rorke LB, Packer RJ et al. Monosomy 22 in rhabdoid

    or atypical tumors of the brain. J Neurosurg 1990;73:710-714.

    44 Burger PC, Yu IT, Tihan T et al. Atypical teratoid/rhabdoid

    tumor of the central nervous system: a highly malignant tumor

    of infancy and childhood frequently mistaken for medulloblas-

    toma: a Pediatric Oncology Group study. Am J Surg Pathol

    1998;22:1083-1092.

    45 Olson TA, Bayar E, Kosnik E et al. Successful treatment of

    disseminated central nervous system malignant rhabdoid

    tumor. Am J Pediatr Hematol Oncol 1995;17:71-75.

    46 Fisher BJ, Siddiqui J, Macdonald D et al. Malignant rhabdoid

    tumor of brain: an aggressive clinical entity. Can J Neurol Sci

    1996;23:257-263.47 Hilden JM, Watterson J, Longee DC et al. Central nervous

    system atypical teratoid tumor/rhabdoid tumor: response to

    intensive therapy and review of the literature. J Neurooncol

    1998;40:265-275.

    48 Oka H, Scheithauer BW. Clinicopathological characteristics of

    atypical teratoid/rhabdoid tumor. Neurol Med Chir (Tokyo)

    1999;39:510-517; discussion 517-518.

    49 Ho DM, Hsu CY, Wong TT et al. Atypical teratoid/rhabdoid

    tumor of the central nervous system: a comparative study

    with primitive neuroectodermal tumor/medulloblastoma.

    Acta Neuropathol (Berl) 2000;99:482-488.

    50 Bhattacharjee M, Hicks J, Langford L et al. Central nervous

    system atypical teratoid/rhabdoid tumors of infancy and

    childhood. Ultrastruct Pathol 1997;21:369-378.51 Versteege I, Sevenet N, Lange J et al. Truncating mutations

    of hSNF5/INI1 in aggressive paediatric cancer. Nature

    1998;394:203-206.

    52 Biegel JA, Zhou J-Y, Rorke LB et al. Germ-line and acquired

    mutations of INI1 in atypical teratoid and rhabdoid tumors.

    Cancer Res 1999;59:74-79.

    53 Kalpana GV, Marmon S, Wang W et al. Binding and stimu-

    lation of HIV-1 integrase by a human homolog of yeast tran-

    scription factor SNF5. Science 1994;266:2002-2006.

    54 Sevenet N, Lellouch-Tubiana A, Schofield D et al. Spectrum of

    hSNF5/INI1 somatic mutations in human cancer and genotype-

    phenotype correlations. Hum Mol Genet 1999;8:2359-2368.

    55 Sevenet N, Sheridan E, Amram D et al. Constitutional muta-

    tions of the hSNF5/INI1 gene predispose to a variety of cancers.

    Am J Hum Genet 1999;65:1342-1348.

    56 Packer RJ, Biegel JA, Blaney S et al. Atypical teratoid/rhab-

    doid tumor of the central nervous system: report on workshop.

    Am J Pediatr Hematol Oncol 2002;24:337-342.

    57 Kalifa C, Hartmann O, Demeocq F et al. High-dose busulfan

    and thiotepa with autologous bone marrow transplantation in

    childhood malignant brain tumors: a phase II study. Bone

    Marrow Transplant 1992;9:227-233.

    58 Mason WP, Grovas A, Halpern S et al. Intensive chemotherapy

    and bone marrow rescue for young children with newly diag-nosed malignant brain tumors. J Clin Oncol 1998;16:210-221.

    59 Slavc I, Schuller E, Czech T et al. Intrathecal mafosfamide

    therapy for pediatric brain tumors with meningeal dissemina-

    tion. J Neurooncol 1998;38:213-218.

    60 Young SW, Qing F, Harriman A et al. Gadolinium(III) texa-

    phyrin: a tumor selective radiation sensitizer that is detectable

    by MRI. Proc Natl Acad Sci USA 1996;93:6610-6615.

    61 Packer RJ, Raffel C, Villablanca JG et al. Treatment of pro-

    gressive or recurrent pediatric malignant supratentorial brain

    tumors with herpes simplex virus thymidine kinase gene vec-

    tor-producer cells followed by intravenous ganciclovir

    administration. J Neurosurg 2000;92:249-254.

    62 Druker BJ, Tamura S, Buchdunger E et al. Effects of a selec-

    tive inhibitor of the Abl tyrosine kinase on the growth of Bcr-

    Abl positive cells. Nat Med 1996;2:561-566.

    63 Huang E, Teh BS, Strother DR et al. Intensity-modulated

    radiation therapy for pediatric medulloblastoma: early report

    on the reduction of ototoxicity. Int J Radiat Oncol Biol Phys

    2002;52:599-605.

    MacDonald, Rood, Santi et al. 186