You are on page 1of 13

J Neurooncol (2009) 92:261273

DOI 10.1007/s11060-009-9829-0

TOPIC REVIEW

The evolution and application of techniques in molecular biology


to human brain tumors: a 25 year perspective
James T. Rutka Paul Kongkham Paul Northcott Carlos Carlotti
Mustafa Guduk Hirokatsu Osawa Orlando Moreno Ho Jun Seol
Andres Restrepo Adrienne Weeks Shoichi Nagai Christian Smith

Received: 1 December 2008 / Accepted: 23 February 2009


Springer Science+Business Media, LLC. 2009

Abstract Since the establishment of the AANS/CNS


Section on Tumors in 1984, neurosurgeons have been
actively involved in basic science research of human brain
tumors that has moved the field forward considerably.
Here, we chronicle the major advances that have been
made with respect to our understanding of the concepts
guiding the biology of human malignant brain tumors.
Numerous technical advances in science, such as the
development of gene transfer techniques, the polymerase
chain reaction, the discovery of oncogenes and tumor
suppressor genes, and the refinement of approaches to
cancer cytogenetics have enabled researchers to identify
many of the non-random genetic alterations associated with
brain tumor growth, invasion, immunology, angiogenesis
and apoptosis. These data led to some astounding progress,
for example with the use of gene therapy, whereby in the
1990s several human clinical trials were conducted for
patients with brain tumors. More recently, the human
genome project has been completed providing a blueprint
for the human species. What has followed are exciting new
techniques in molecular biology such as transcriptional
profiling, single nucleotide polymorphism (SNP)-arrays,
array comparative genomic hybridization (array-CGH),
microRNA profiling, and detection of epigenetic silencing
of tumor suppressor genes. The cancer genome is now
being sequenced at break neck speed using advanced DNA
sequencing techniques. We are on the threshold of
J. T. Rutka (&)  P. Kongkham  P. Northcott  C. Carlotti 
M. Guduk  H. Osawa  O. Moreno  H. J. Seol  A. Restrepo 
A. Weeks  S. Nagai  C. Smith
The Arthur and Sonia Labatt Brain Tumour Research Centre,
Division of Neurosurgery, The Hospital for Sick Children, The
University of Toronto, Suite 1503, 555 University Avenue, M5G
1X8 Toronto, ON, Canada
e-mail: james.rutka@sickkids.ca

cataloguing the major genetic alterations observed in all


human brain tumors. What will follow is modeling of these
genetic alterations in systems that will allow for the
development of novel pharmacotherapeutics and translational research therapies.
Keywords Brain tumor  Molecular biology 
Cytogenetics  Epigenetics  MicroRNAs 
DNA sequencing

Introduction
Patients with malignant brain tumors continue to pose
tremendous challenges to their treating neurologists, neurosurgeons, radiation oncologists, and neuro-oncologists.
Twenty-five years ago, our understanding of the basic
biology of brain tumors was in its infancy. There was
arguably little progress that could be claimed then that
would impact favorably on the course of the disease for
most patients. However, in the short course of 25 years, our
knowledge base for these tumors has increased dramatically. At least some of the victories that we can now claim
for patients with malignant brain tumors arise out of a
better characterization of the signaling and cell cycle
pathways, cell surface markers, tumor microenvironment,
and genetic and epigenetic disturbances that comprise these
tumors. Here, we chronicle the formidable advances that
have occurred through the application of techniques in
molecular biology to the study of human brain tumors over
the past 25 years. It is probably fair to say that since the
creation of the AANS/CNS Section on Tumors in 1984, the
techniques used to probe the most fundamental aspects of
these tumors have advanced at a faster rate than has
ever occurred before in history. For example, whereas in

123

262

1984 the genetic alterations in human brain tumors were


determined through conventional G-banding karyotyping
(Fig. 1), and required a skilled cytogeneticist for interpretation, today comprehensive genome wide surveys and
sequencing can be performed which sift through billions of
datapoints to arrive at the key genetic disturbances that
characterize human cancer.

A look into the past: brain tumor research


in the 1980s
In the early 1980s, streams of advancements in genetics and
molecular biology were converging to provide new tools
with which research investigators could ask questions
regarding the etiology of human brain tumors. Among these
tools were monoclonal antibodies which could be used to
detect the presence or quantity of a protein in tumor cell
lysates using Western blot analysis or in fixed and preserved
tumor specimens using immunohistochemistry. The promise of monoclonal antibodies was exciting for if brain
tumors were found to express unique antigens which were
not expressed by normal cells, then antibodies could be
modified so as to deliver a toxin, radio-isotope or cytokine
selectively to a brain tumor cell. This line of investigation
has been pursued vigorously by numerous researchers over
the years. Some of the best examples include the work by
Bigner and colleagues who determined that tenascin is
overexpressed by human malignant gliomas and against
which monoclonal antibodies were generated [1, 2]. These
antibodies, labeled with radio-isotopes, have now been used
in clinical trials in patients harboring malignant gliomas
with significant efficacy [39].
Just as the field of monoclonal antibodies and immunotherapy was developing in the early 1980s, the recognition
Fig. 1 Classic G-banding
karyotyping of a
medulloblastoma is seen in
panel (D). CGH of this tumor
reveals gains on chromosomes 5
and 7, panel (A). SKY paints are
applied to the tumor providing
color coding of each
chromosome, panels (B) and
(C). Translocations are readily
seen in the SKY metaphase
preparation of this
medulloblastoma involving
chromosomes 3 and 7; 10 and
14; and 7q and 11 q, panel (E).
From Bayani et al. [31] with
permission

123

J Neurooncol (2009) 92:261273

of latent cancer causing genes in the human genome, socalled oncogenes was quickly becoming appreciated. In
1976, Bishop and Varmus demonstrated that oncogenes
were defected proto-oncogenes that are found in several
species including humans [10]. Proto-oncogene activation
may be caused by point mutations, chromosomal translocations, deletions and gene amplifications [11]. Among
some of the oncogenes that were identified in human brain
tumors at that time were c-myc, N-ras, v-sis, and v-fos for
human malignant gliomas [12].
It was in the 1980s that the hypothesis that growth
factors, growth factor receptors, and their related biochemically active secondary and tertiary messages was
rapidly gaining momentum. It was hypothesized that normal cellular proliferation could be disrupted, and malignant
transformation could occur if a growth factor cascade was
altered by, for example, overexpression of a particular
growth factor or growth factor receptor [12]. Some of the
growth factor activation pathways that were studied in
human brain tumors included platelet-derived growth factor
(PDGF), epidermal growth factor (EGF), and fibroblast
growth factor (FGF). Research studies from this time led to
the discovery that human glioblastoma multiforme (GBM)
could be characterized by amplification and overexpression
of the epidermal growth factor receptor (EGFR) in as many
as 30% of cases [1315].
While investigators were beginning to analyze human
brain tumors for their expression of oncogenes, growth
factors, and growth factor receptors, a number of technical
advancements in molecular biology came forward to speed
up this process. They included the routine application of
restriction endonucleases to biological samples to reduce
large pieces of DNA into smaller, more manageable fragments; the development of the polymerase chain reaction
(PCR) to make large number copies of specific DNA

J Neurooncol (2009) 92:261273

fragments or genes for sequencing and further analysis; the


widespread use of DNA transfer technology which enabled
genes to be expressed ectopically in model systems and
which foreshadowed the development of gene therapy (see
next section); and the improvement of DNA sequence
technology to identify genes with greater reliability and
speed that are involved in human cancer. In 1988, the
National Center for Biotechnology Information (NCBI) was
founded to create a public database wherein biomedical
information and gene sequencing data are made available
for the better understanding of molecular processes affecting human health and disease, including cancer.

Harnessing the energy of molecular biology towards


human brain tumors: progress in the 1990s
One of the most exciting lines of investigation in the 1990s
relating to brain tumors was the use of gene therapy as a
means to control the growth and invasion of these tumors.
Gene therapy is defined as the use of nucleic acid transfer,
either RNA or DNA, to treat or prevent disease [16, 17].
Success in gene therapy requires an understanding of
disease pathogenesis, the ability to specifically target the
tissue of interest, and the elucidation of an effective therapeutic gene(s) in addition to an appropriate animal model to
study (reviewed in [18]). Glioblastoma was thought to be a
particularly good candidate for cancer gene therapy given
the rapid cellular proliferation associated with this tumor in
the context of normal postmitotic tissue, facilitating the use
of delivery vehicles or target molecules specific for dividing
cells. Pioneering work in gene therapy of malignant gliomas
involved direct injection of herpes simplex virus (HSV)
lacking the thymidine kinase (TK) gene [19]. Potent antitumor effects were noted in both established glioma cell
lines and short-term populations of glioma cells following
treatment with TK-negative HSV. Ram et al. [20] reported
that antitumor effects following implantation of retroviral
(RV) producer cells were observed only in small tumors
experimentally (\1.5 ml). Palu et al. [21] evaluated RV
delivery of IL-2 in combination with HSV-TK therapy in a
clinical trial. Follow-up of patients was brief but all four
patients were shown to have reduced tumor volume. In
essence, gene therapy seemed most promising at this time,
but subsequent studies alluded to some of the difficulties
with this technique including tumor targeting, enhanced
immunogenicity of constructs, and concern about systemic
spread of virus. The fact that human trials were conducted
with adequate safety and without significant complications
is a testament to the power of this technique. The interested
reader is referred to the results of a number of human gene
therapy trials that were conducted in the 1990s for patients
with malignant gliomas [2026].

263

It was because of the rapid advances in techniques in


molecular biology in the 1980s that the US Department
of Energy and the National Institutes of Health launched
the human genome project (HGP) in 1990. While the
purpose of the HGP was to identify all genes in human
DNA, the knowledge derived from these data would
unquestionably help in the study of diseases, such as
cancer, where the direct DNA sequence may be perturbed. Numerous strategies then evolved which enabled
investigators to interrogate the cancer genome with more
precision than ever before. As a result, the field of
cancer cytogenetics developed to help researchers discover the main genetic aberrations that occur in human
brain tumors.
Fluorescein in situ hybridization (FISH) techniques
arose when molecular probes could be synthesized against
unique DNA sequences so as to visualize the gene
sequence in a given cell. FISH is used to identify numerical
and structural chromosomal abnormalities, detect minimal
residual disease, and to map gene locations. For example,
FISH is an excellent technique to identify losses of chromosome 1p and 19q in human oligodendroglioma [27], or
to see loss of chromosome 22q11 in atypical teratoid
rhabdoid tumors (ATRTs) [28].
Spectral karyotyping (SKY) was first reported in 1996,
and uses optical spectroscopy and chromosomal paints to
examine the orderly arrangement of all the human chromosomes. SKY can detect the translocation of a DNA fragment
as small as 1.5 million base pairs, and can identify the
chromosome of origin of various extrachromosomal fragments. SKY has been particularly useful in neuro-oncology
in tumors where translocations have been frequently identified, such as Ewings Sarcoma (ES) in children. Intracranial
ES can be diagnosed with accuracy by SKY through the
identification of a characteristic t(11;22)(q24:q12) translocation seen in 85% of tumors [29].
A variation on FISH technology is comparative genomic
hybridization (CGH) which, unlike FISH, allows for the
detection of changes of a few hundred kilobase pairs. CGH
quantitate the ratio of two differently colored fluorescence
probes in an in situ hybridization experiment. CGH has
been used to confirm, for example, that primary GBMs
exhibit a high frequency of chromosomal gains compared
to secondary GBMs [30].
In one of our studies, we combined the use of Giemsabanding, comparative genomic hybridization, and spectral
karyotyping to examine the genetic alterations in human
medulloblastoma. Here we found that the combination of
these three techniques led to the determination of the
greatest number of cytogenetic alterations in medulloblastoma including rearrangements on chromosomes 7 and
17, amplification on chromosome 2q in 30% of cases, and
loss of chromosome 10q (Fig. 1) [31].

123

264

In the 1990s a powerful technique in molecular biology


emerged which enabled the detection of over- or underexpressed genes in brain tumors when compared to control,
non-neoplastic specimens. This technique is called cDNA
microarray or transcriptional profiling analysis, and uses
thousands of nucleic acid probes to interrogate the
expression of thousands of genes in a given sample. New
generation high-resolution arrays have now been developed
which interrogate every annotated human exon. In addition, the creation of genome tiling arrays enable the
measurement of gene expression at every 25 base pairs
across the entire genome so that even cryptic transcriptional units can be uncovered [32]. Transcriptional
profiling has now been performed on numerous brain tumor
subtypes such that novel genes and pathways have been
identified and further studied to increase our knowledge of
the origins of these tumors (Fig. 2) [3338]. Several years
ago, we used the Atlas 1.2 human cancer array platform to
determine genes that were overexpressed in medulloblastoma when compared to normal human cerebellum. Our
analysis led to the identification of 6 genesezrin, cyclin
D2, high mobility group protein 2, MAPRE1, histone
deacetylase 2 and ornithine decarboxylase 1which were
overexpressed several fold in medulloblastoma. From this
list of genes, we pursued the role of ezrin in medulloblastoma, and determined that its overexpression greatly
enhanced the metastatic potential of medulloblastoma in an
orthotopic xenograft model system [39].

The new millennium: oncogenomics, and epigenetic


alterations in human brain tumours
Recent advances in oncogenomics
The new millennium will be characterized by phenomenal
progress in cataloguing the multitude of genetic aberrations
that characterize human brain tumors. Numerous new
techniques are advancing the field of oncogenomics at
record breaking pace. Array CGH is one such technique,
and is similar to CGH. However, instead of hybridizing the
labeled probes to metaphase chromosomes which is done
in standard CGH cases, in array CGH the probes are
hybridized to microarray slides in which thousands of
fragments of DNA that span the genome have been
arrayed out [40]. Array CGH has been used to study a
wide variety of human brain tumors [4144]. Taylor et al.
[44] used array CGH and transcriptional profiling to show
that histologically similar ependymomas arising from different regions of the neuraxis had markedly different
patterns of copy-number-driven gene expression. Rossi
et al. [43] used array CGH in pediatric medulloblastoma to
show that loss of 17p and gain of 17q are the most common

123

J Neurooncol (2009) 92:261273

cytogenetic alterations in medulloblastoma, and that


amplifications are rare with the most common one being
the MYC gene on chromosome 8q.
One of the most significant advances in cancer genomics
has been the development of single nucleotide polymorphism (SNP) arrays. SNPs are naturally occurring variations
in DNA sequence that occur on average at one SNP for
every 200 bases of DNA, and are the most common form of
genetic variation in the human genome. Modern SNP array
platforms interrogate from 100,000 to 2,000,000 SNPs
resulting in high resolution genome coverage with median
SNP spacing of approximately 7001500 base pairs. This
resolution is more than 1,000 times better than classic
CGH. Recently, a comprehensive genomic characterization
was performed on 206 glioblastoma multiformes using
genomic copy number alterations (CNAs) on three microarray platforms including SNP-based arrays [45]. This study
showed high level amplifications of EGFR, CDK4, and
PDGFRA, and homozygous deletions of CDKN2A/B and
PTEN among many other findings. We have used SNP array
platforms to query over 250 primary medulloblastoma
samples, and have uncovered numerous target regions of
interest (Fig. 2) [46].
Beyond oncogenomics: cancer epigenetics
comes of age
Until recently, cancer researchers were focused primarily
on the discovery of tumor suppressor genes that were
inactivated by gene mutations and/or deletions. Now we
know that epigenetic factors can regulate gene expression,
and can lead to tumor suppressor gene silencing (Fig. 3).
Epigenetics refers to mechanisms controlling gene
expression in a heritable fashion, without any changes to
the gene sequence itself. Epigenetic mechanisms including
promoter methylation, histone modifications, and microRNAs (miRNAs) (see below) function to regulate normal
processes such as gene imprinting, X-chromosome inactivation, and developmental stage- and tissue-specific gene
expression (Fig. 4). Promoter methylation involves the
covalent addition of a methyl group to cytosine bases found
in CG dinucleotide-rich regions of the genome referred to
as CG islands. CG islands are found at the promoter
regions in approximately 60% of human genes. In normal
tissues, these CG islands are typically unmethylated,
whereas in cancer they may become extensively methylated (Fig. 5). Promoter methylation is generally associated
with transcriptional silencing, resulting from the inhibition
of transcription factor binding or the recruitment of transcriptional repressor molecules (Fig. 6) [47]. Histones are
protein complexes involved in packaging DNA into chromatin within the nucleus. The N-terminal tails of histone
proteins are subject to a myriad of post-translational

J Neurooncol (2009) 92:261273

265

Fig. 2 a Isochromosome 17q in medulloblastoma. High-resolution


SNP arrays are an efficient platform for detecting copy number
aberrations in tumour samples. Shown is the copy number output for
chromosome 17 in ten medulloblastomas, with losses depicted in blue
and gains in red. The most common structural abnormality in
medulloblastoma, isochromosome 17q, is observed in the majority of
the cases, as evidenced by the characteristic loss of 17p with

concomitant gain of 17q. b Gene expression profile of significant


genes on chromosome 17 in medulloblastoma. Oligonucleotide
expression arrays are a sensitive method for the detection of
differentially expressed genes in tumour samples. The heat map
highlights the most variable genes on chromosome 17 in a series of 7
medulloblastomas. Genes with reduced expression are shown in green
and those with elevated expression are red

modifications, including methylation and acetylation. In


general, acetylation is associated with a transcriptionally
competent euchromatin structure whereas deacetylation
results in a transcriptionally repressed heterochromatic
configuration. Histone methylation can be associated with
gene expression or silencing, depending on the specific
histone protein and lysine residue targeted by this modification. Taken together, the various histone modifications
have been referred to as the histone code [48]. Micro
RNAs are a group of small, non-coding RNAs involved in
regulating gene expression, and are discussed separately in
the next section.
Dysregulation of these epigenetic mechanisms may cause
altered gene expression (e.g., silencing of tumor suppressor
genes), contributing to the etiology of human cancer.
Abnormal promoter methylation or histone modifications

may confer a stable selective growth advantage on a cell


population, resulting in clonal expansion and tumorigenesis.
In some instances, epigenetic dysregulation may play an
early role in cancer initiation, with aberrant silencing of
epigenetic gatekeeper genes allowing for inappropriate
survival and expansion of a population of cells at risk of
acquiring additional genetic defects [49].
Over the last two decades there has been increasing
interest in the role epigenetic dysregulation plays, alongside genetic events, in the etiology of human brain tumors.
To date, the influence of epigenetic phenomena in brain
tumors has probably been most studied in malignant gliomas and pediatric medulloblastomas. Several classic tumor
suppressor genes, including PTEN, p53 and p14/ARF have
been shown to be silenced epigenetically in human gliomas
[5052]. Genome-wide epigenomic profiling of malignant

123

266

J Neurooncol (2009) 92:261273

Mutation

Deletion

CH 3

Epigenetics

CH 3

Fig. 3 Tumor suppressor genes may be silenced due to genetic


events such as mutation or deletion, or epigenetic events including
DNA methylation or histone modifications

Histone
Modifications

DNA
Methylation

GENE SILENCING

miRNA

Fig. 4 Epigenetic gene silencing. Several epigenetic mechanisms


cooperate to produce gene silencing, including DNA promoter
methylation, histone modifications, and micro-RNA (miRNA) mediated gene silencing. If gene silencing occurs at the locus of a tumor
suppressor gene, then tumor formation can occur

glioma cell lines and primary tumor samples has identified


a number of additional candidate genes targeted for aberrant silencing, including CST6, BIK, TSPYL5, BEX1, and

123

BEX2 [53, 54]. Deregulation of the BMP signaling pathway resulting from promoter methylation-mediated
silencing of the BMP receptor 1B has been implicated in
glioma tumor stem cell tumorigenicity [55]. In addition to
its utility in understanding the pathogenesis of human brain
tumors, the study of epigenetic gene silencing may serve
prognostic purposes, as highlighted recently by the association between O(6)-methylguanine methyltransferase
(MGMT) promoter methylation and improved progressionfree and overall survival in malignant glioma patients
treated with oral alkylating chemotherapeutic agents [56].
Additional novel epigenetic prognostic markers are also
being explored [57].
Among pediatric brain tumors, medulloblastoma has
been the most intensely studied tumor type with respect to
the contribution of epigenetic phenomena to its etiology.
Strong evidence for promoter methylation-mediated epigenetic silencing in MB exists for HIC1 and RASSF1A,
with additional evidence available for a small number of
genes including CASP8, MGMT, GSTP1, p14, p15, p16,
and TIMP3 [58]. We recently showed that SPINT2, a
negative regulator of the HGF-Met signaling pathway, is
silenced by methylation in approximately 30% of primary
medulloblastomas [59]. The WNT signaling inhibitor,
DKK1, has been shown to be silenced secondary to inappropriate histone modifications in medulloblastoma [60].
MicroRNAs: small molecules with big function
MicroRNAs (miRNAs) belong to a recently described class
of small regulatory RNAs that coordinate important cellular processes, including proliferation and differentiation.
Since the 1993 discovery of lin-4 in C. elegans [61, 62],
thousands of miRNA sequences have been annotated in a
vast number of metazoan, plant, and viral genomes. With
695 unique miRNAs currently reported in humans [63], it
is estimated that they account for [3% of all human genes
[64]. As this inventory of known miRNAs continues to
increase, it is becoming increasingly evident that these
non-coding RNAs function to regulate a variety of cellular
processes including development, differentiation, proliferation, and apoptosis [64]. At present, miRNAs are probably
best described for their role in developmental processes,
most of which have been established by studies in C. elegans and Drosophila [65, 66]. In addition, several recent
reports have provided compelling evidence that deregulation of miRNAs contributes to the pathogenesis of human
disease, particularly cancer [6769].
Mature miRNAs consist of *22 nucleotide, singlestranded RNA molecules which function as a component of
the miRISC (miRNA-containing RNA-induced silencing
complex) to post-transcriptionally regulate target mRNAs
sharing sequence complementarity in their 30 untranslated

J Neurooncol (2009) 92:261273


Fig. 5 Detection of gene
promoter methylation by
bisulfite sequencing. Promoter
methylation status in normal
tissue versus tumor tissue.
Typically, CG islands remain
unmethylated (open circles) in
normal neural tissues. In brain
tumors, CG islands may become
extensively methylated,
resulting in abnormal gene
silencing (filled circles).
Vertical tick marks denote CG
sites within the promoter CG
island

267

-500
EXON 1

Normal Brain

Brain Tumor

Unmethylated CG Site

Fig. 6 In normal tissue,


promoter CG islands are largely
unmethylated, resulting in a
transcriptionally competent
euchromatin state. When these
CG islands become methylated,
transcription is abolished, due to
the conversion to a closed
heterochromatin state, the
prevention of transcription
factor binding, or the
recruitment of transcriptional
repressors

Normal

Methylated CG Site

RNA

TF

TF

RNA Pol

Aberrant Promoter
CG Island Methylation

Brain Tumor
RNA Pol

TF
TF

Transcription
Abolished

Unmethylated CG Site

Methylated CG Site

123

268

J Neurooncol (2009) 92:261273

years, there is abundant evidence to support miRNAs acting


as oncogenes and tumor suppressors [68, 69]. An association between miRNAs and human cancer was first reported
in 2002 when it was shown that miR-15a and miR-16a map
to chromosome 13q14, a region deleted at high frequency in
chronic lymphocytic leukemia (CLL) [71]. A follow-up
study published by the same group revealed that miRNAs
are commonly located at fragile sites and other cancerassociated regions in the human genome [72]. These findings were the first indication of what has very recently
become a prevailing theme in the miRNA literature: miRNAs constitute a new class of oncogenes and tumour
suppressors.
The best characterized oncogenic miRNAs thus far
identified are components of the polycistronic mir-17-92
cluster, which maps to chromosome 13q31 and produces six
mature miRNAs [73]. This locus is recurrently amplified in
follicular lymphoma, B-cell lymphoma, and several lung,
and head and neck cancers [74, 75]. In addition, direct
evidence for this miRNA cluster acting as an oncogene has
been shown in a mouse model of B-cell lymphoma, where a
truncated portion of miR-17-92 (miR-17-19b) was capable
of cooperating with c-Myc overexpression to accelerate
lymphomagenesis [75]. Additional miRNAs have also been

regions (UTR) (Fig. 7). Target mRNAs are either degraded


or repressed translationally by the cognate miRNA,
depending on the degree of complementarity that exists
between the miRNA and the target. The short nature of
mature miRNAs and the somewhat liberal requirements for
target recognition make it possible for a single miRNA to
regulate possibly hundreds of mRNAs [63]. With the
number of predicted targets that are possible for a given
miRNA, it is not difficult to imagine how a limited number
of miRNAs may have the capacity to effectively impact
cellular phenotype. This possibility was supported in an
elegant study by Lim and colleagues [70] who demonstrated that a single miRNA could cause downregulation of
a large number of mRNAs (*100) and shift the expression
profile of miRNA-transfected cells to mirror that of the
specific tissue in which the miRNA is normally expressed.
It can be appreciated from this report how disruption in the
normal expression pattern of miRNAs might have detrimental effects on the cellular environment and, in turn,
contribute to the initiation and progression of malignancy.
The discovery of miRNAs as key post-transcriptional
regulators of the cells transcriptome has led to an intense
amount of research aimed at the characterization of these
molecules in tumorigenesis [67, 68]. Indeed, in the last few

rosha
Drosha

Pre-miRNA
nt)
re-miRNA (~80
~80 nt

Pri-miRNA
Pri-miRN

miRNA gene

Nucleus
Dicer
cer

Cytoplasm

Mature miRNA (~22 nt)

Translational repression
m7GpppG
Target mRNA

5 UTR

ORF

3 UTR

AAAAA

Imperfect
complementarity

Fig. 7 Schematic of miRNA biogenesis. miRNAs are initially


transcribed as long primary transcripts (pri-miRNAs) by RNA
polymerase II, before being processed by Drosha in the nucleus to
generate pre-miRNAs (*80 nt) which form a characteristic stemloop conformation. Pre-miRNAs are exported from the nucleus into
the cytoplasm where they are further processed by Dicer to form the

123

mRNA degradation
m7GpppG
Target mRNA

5 UTR

ORF

3 UTR

AAAAA

Perfect
complementarity

mature miRNA (*22 nt). Mature miRNAs post-transcriptionally


regulate target gene expression by forming hybrid complexes with
target mRNAs in their 30 untranslated region (UTR), and subsequently
repress translation or promoting degradation of the target mRNA,
depending on the amount of complementarity that exists between the
miRNA and its target(s)

J Neurooncol (2009) 92:261273

implicated as oncogenes based upon there expression profiles in human cancer, including miR-155 which has been
reported to be aberrantly expressed in B-cell lymphoma
[76] and miR-21 which has been shown to be upregulated in
a variety of solid tumours [77, 78].
Of the miRNAs currently believed to represent bona fide
tumour suppressors, let-7 is likely the most prominent.
Expression of let-7 has been reported to be significantly
reduced in human lung cancers [79]. The significance of
this observation is underscored by the finding that the let-7
family is known to regulate Ras [80], suggesting a mechanism by which reduced let-7 expression could result in
activation of a potent oncogene.
A number of miRNAs have recently been associated with
tumours of the central nervous system. For example, miR-21
is frequently upregulated in high-grade gliomas [77, 81].
MicroRNA expression profiling showed that miR-21 was
consistently overexpressed in glioblastomas and established
glioblastoma cells lines compared to non-neoplastic control
cells derived from fetal and adult brain [77, 81]. Additionally, knockdown of miR-21 in glioblastoma cells leads to
caspase activation and increased apoptosis in vitro and is
able to synergize with neural precursor delivered S-TRAIL
to inhibit glioma cell proliferation and survival in vivo [81].
In contrast to miR-21 overexpression, several miRNAs
have been implicated as tumour suppressors in brain tumors
as a result of their observed downregulation. miR-124 and
miR-137 have been reported to exhibit reduced expression in
anaplastic astrocytomas and glioblastoma multiforme. Their
exogenous expression in either normal neural or brain tumor
stem cells promotes morphological and expression changes
consistent with cellular differentiation [82]. miR-124
expression has also been shown to be reduced in
medulloblastomas and re-expression of miR-124 in a
medulloblastoma cell line resulted in downregulation of the
CDK6 oncogene and slowed proliferation [83]. A very recent
report identified miR-128 as significantly downregulated in
primary glioblastomas compared to adjacent normal tissue
devoid of tumor cells [84]. This study implicated miR-128 as
a putative tumor suppressor in glioblastoma capable of
repressing the oncogenic properties of Bmi1 and inhibiting
self-renewal of glioma stem-like cells [84].
We have recently shown that the miRNA-17/92 polycistron is amplified and upregulated in Sonic Hedgehog-driven
medulloblastomas, and is induced by N-Myc in Sonic
Hedgehog-treated cerebellar neural precursor cells [85].

Back to the future: advanced cancer genome


sequencing
Recent technical breakthroughs in DNA sequencing platforms and their utility in genomic analyses have resulted in

269

a number of coordinated efforts focused on comprehensive


characterization of the cancer genome. Indeed The Cancer
Genome Atlas (TCGA), led by the National Cancer Institute
and the National Human Genome Research Institute, is a
multi-institutional consortium aimed at improving our
understanding of cancer through genomic characterization
and high-throughput next-generation sequencing technologies. The first study published by the TCGA involved
integrated DNA copy number, gene expression, and DNA
methylation profiles of 206 glioblastomas [45]. PCRdirected exon re-sequencing was performed for 601 selected
genes in 91 of the tumours. This approach of integrating
multi-dimensional genomic data from glioblastomas
revealed the consistent deregulation of p53, RB, and PI(3)
kinase signaling pathways in the majority of tumors and
established a connection between MGMT promoter methylation and a hypermutator phenotype in some cases [45]. In
a similar study, 22 glioblastomas were comprehensively
profiled by exon re-sequencing of 20,661 protein-coding
genes, analyzed for copy number aberrations using SNP
arrays, and gene expression by combining SAGE (Serial
Analysis of Gene Expression) with next-generation
sequencing [86]. This report identified recurrent mutations
in IDH1 in 12% of GBM patients, mostly in young patients
and those with secondary GBMs, and showed that patients
harboring IDH1 mutations had increased overall survival.
Although both of the studies mentioned above relied
primarily on PCR-based Sanger-sequencing, many groups
are now taking advantage of next-generation/next-gen or
deep sequencing methods for studying normal and disease genomes. There are currently three main next-gen
platforms on the market: 454 (Roche), Solexa (Illumina),
and SOLiD (Applied Biosystems). 454 and Solexa are
based on sequencing by synthesis chemistries and SOLiD
involves sequencing by ligation (Reviewed in [87]).
Despite differences in sample preparation, sequencing
chemistry, and read length, the end result is up to tens of
gigabases of sequencing data from a single run.
The high-throughput afforded by next-gen sequencing
has recently enabled several significant breakthroughs in
genomics. For example, when combined with array-mediated exon capture technology, targeted re-sequencing of
*200,000 protein-coding exons in a single experiment has
been achieved [88]. In addition, using a paired-end strategy
to generate sequence reads from both ends of millions of
DNA fragments, Campbell et al. [89] identified [100
somatically acquired genomic rearrangements in human
lung cancer cell lines. Other studies have exploited next-gen
sequencing to identify transcription factor targets and map
histone modifications using ChIP-Seq [9092] and measure
gene expression profiles and detect alternative splicing
using mRNA-Seq [93, 94]. It can be anticipated that many
of these novel sequencing-based methods will be routinely

123

270

J Neurooncol (2009) 92:261273

applied in cancer genomics, including the study of brain


tumours in the near future.

Children, and B.r.a.i.n child. Paul Kongkham is a recipient of a


National Cancer Institute of Canada Terry Fox Foundation research
fellowship.

Conclusions

References

How are these molecular applications influencing patient


diagnosis and treatment today? As some examples, the loss
of 1p and 19q in anaplastic oligodendrogliomas affords
patients a survival advantage while on temozolomide
chemotherapy; methylation of the MGMT promoter is predictive of response to temozolomide and radiation therapy
for patients with glioblastoma multiforme; loss of 22q11 by
FISH in a posterior fossa tumor in a child establishes the
diagnosis of ATRT; and the presence of nuclear beta-catenin
by immunohistochemistry in a childhood medulloblastoma
is associated with an excellent long term prognosis.
As new genetic disturbances are uncovered through
advanced cancer genome sequencing, these gene alterations will be modeled in rodents and subhuman primates
before their translation to the clinic. These models offer an
opportunity for delivery of novel pharmacotherapeutics for
these tumors. Once efficacy is established, these new
agents can be taken to human patients harboring brain
tumors as translational therapies. Such has certainly been
the case for other cancer subtypes, such as breast and lung,
where small molecule inhibitors of known signaling pathways have been developed to treat patients in clinical trials.
Examples of these include Bevacizumab (Avastin) as an
anti-angiogenic agent targeting the VEGF pathway which
is now being tested in patients with gliomas; Sirolimus
(Rapamycin) which targets the mTOR pathway and is
being used to halt tumor growth in patients with tuberous
sclerosis and neurofibromatosis; and CDX-110, a vaccine
trial in glioma patients in whom a vaccination directed at
an EGFR-mutant is being used in clinical trials.
The chronicling here of the many advances in brain
tumor diagnosis and treatment with the use of applications
in molecular biology these past 25 years suggests that the
future will be characterized by equal change and
advancements. However, the translation of our understanding of the molecular biology and genetics of human
brain tumors into a tangible benefit to the patient has
remained elusive despite all the sophisticated molecular
genetic platforms and tools that are available. Hence, we
are cautiously optimistic that we are proceeding along the
appropriate pathways and channels to improve the survival
and quality of life of patients with malignant brain tumors
over the next 25 years.

1. Bourdon MA, Matthews TJ, Pizzo SV, Bigner DD (1985)


Immunochemical and biochemical characterization of a gliomaassociated extracellular matrix glycoprotein. J Cell Biochem
28:183195. doi:10.1002/jcb.240280302
2. Bourdon MA, Wikstrand CJ, Furthmayr H, Matthews TJ, Bigner
DD (1983) Human glioma-mesenchymal extracellular matrix
antigen defined by monoclonal antibody. Cancer Res 43:2796
2805
3. Bigner DD, Brown M, Coleman RE, Friedman AH, Friedman
HS, McLendon RE, Bigner SH, Zhao XG, Wikstrand CJ, Pegram
CN et al (1995) Phase I studies of treatment of malignant gliomas
and neoplastic meningitis with 131I-radiolabeled monoclonal
antibodies anti-tenascin 81C6 and anti-chondroitin proteoglycan
sulfate Me114 F (ab)2a preliminary report. J Neurooncol
24:109122. doi:10.1007/BF01052668
4. Cokgor I, Akabani G, Kuan CT, Friedman HS, Friedman AH,
Coleman RE, McLendon RE, Bigner SH, Zhao XG, GarciaTurner AM, Pegram CN, Wikstrand CJ, Shafman TD, Herndon
JEII, Provenzale JM, Zalutsky MR, Bigner DD (2000) Phase I
trial results of iodine-131-labeled antitenascin monoclonal antibody 81C6 treatment of patients with newly diagnosed malignant
gliomas. J Clin Oncol 18:38623872
5. Colapinto EV, Lee YS, Humphrey PA, Zalutsky MR, Friedman
HS, Bullard DE, Bigner DD (1988) The localisation of radiolabelled murine monoclonal antibody 81C6 and its Fab fragment in
human glioma xenografts in athymic mice. Br J Neurosurg
2:179191. doi:10.3109/02688698808992668
6. Lee Y, Bullard DE, Humphrey PA, Colapinto EV, Friedman HS,
Zalutsky MR, Coleman RE, Bigner DD (1988) Treatment of
intracranial human glioma xenografts with 131I-labeled antitenascin monoclonal antibody 81C6. Cancer Res 48:29042910
7. McLendon RE, Akabani G, Friedman HS, Reardon DA, Cleveland
L, Cokgor I, Herndon JEII, Wikstrand C, Boulton ST, Friedman
AH, Bigner DD, Zalutsky MR (2007) Tumor resection cavity
administered iodine-131-labeled antitenascin 81C6 radioimmunotherapy in patients with malignant glioma: neuropathology
aspects. Nucl Med Biol 34:405413. doi:10.1016/j.nucmedbio.
2007.01.009
8. Reardon DA, Akabani G, Coleman RE, Friedman AH, Friedman
HS, Herndon JEII, Cokgor I, McLendon RE, Pegram CN, Provenzale JM, Quinn JA, Rich JN, Regalado LV, Sampson JH,
Shafman TD, Wikstrand CJ, Wong TZ, Zhao XG, Zalutsky MR,
Bigner DD (2002) Phase II trial of murine (131)I-labeled antitenascin monoclonal antibody 81C6 administered into surgically
created resection cavities of patients with newly diagnosed
malignant gliomas. J Clin Oncol 20:13891397. doi:10.1200/
JCO.20.5.1389
9. Rizzieri DA, Akabani G, Zalutsky MR, Coleman RE, Metzler
SD, Bowsher JE, Toaso B, Anderson E, Lagoo A, Clayton S,
Pegram CN, Moore JO, Gockerman JP, DeCastro C, Gasparetto
C, Chao NJ, Bigner DD (2004) Phase 1 trial study of 131I-labeled
chimeric 81C6 monoclonal antibody for the treatment of patients
with non-Hodgkin lymphoma. Blood 104:642648. doi:10.1182/
blood-2003-12-4264
10. Stehelin D, Varmus HE, Bishop JM, Vogt PK (1976) DNA
related to the transforming gene(s) of avian sarcoma viruses is
present in normal avian DNA. Nature 260:170173. doi:10.1038/
260170a0

Acknowledgments This work was supported by grants from the


Canadian Institutes of Health Research (MOP-74610), the Pediatric
Brain Tumor Foundation, the Wiley Fund at the Hospital for Sick

123

J Neurooncol (2009) 92:261273


11. Bishop JM (1983) Cellular oncogenes and retroviruses. Ann Rev
biochem 52:301354
12. Rutka JT, Trent JM, Rosenblum ML (1990) Molecular probes in
neuro-oncology: A Review. Cancer Invest 8:425438. doi:
10.3109/07357909009012060
13. Libermann TA, Nusbaum HR, Razon N, Kris R, Lax I, Soreq H,
Whittle N, Waterfield MD, Ullrich A, Schlessinger J (1985)
Amplification, enhanced expression and possible rearrangement
of EGF receptor gene in primary human brain tumours of glial
origin. Nature 313:144147. doi:10.1038/313144a0
14. Libermann TA, Nusbaum HR, Razon N, Kris R, Lax I, Soreq H,
Whittle N, Waterfield MD, Ullrich A, Schlessinger J (1985)
Amplification and overexpression of the EGF receptor gene in
primary human glioblastomas. J Cell Sci Suppl 3:161172
15. Wong AJ, Bigner SH, Bigner DD, Kinzler KW, Hamilton SR,
Vogelstein B (1987) Increased expression of the epidermal
growth factor receptor gene in malignant gliomas is invariably
associated with gene amplification. Proc Natl Acad Sci USA
84:68996903. doi:10.1073/pnas.84.19.6899
16. Crystal RG (1995) Transfer of genes to humans: early lessons and
obstacles to success. Science 270:404410. doi:10.1126/science.
270.5235.404
17. Mulligan RC (1993) The basic science of gene therapy. Science
260:926932. doi:10.1126/science.8493530
18. Robbins PD, Ghivizzani SC (1998) Viral vectors for gene therapy. Pharmacol Ther 80:3547. doi:10.1016/S0163-7258(98)
00020-5
19. Martuza RL, Malick A, Markert JM, Ruffner KL, Coen DM
(1991) Experimental therapy of human glioma by means of a
genetically engineered virus mutant. Science 252:854856. doi:
10.1126/science.1851332
20. Ram Z, Culver KW, Oshiro EM, Viola JJ, DeVroom HL, Otto E,
Long Z, Chiang Y, McGarrity GJ, Muul LM, Katz D, Blaese RM,
Oldfield EH (1997) Therapy of malignant brain tumors by intratumoral implantation of retroviral vector-producing cells. Nat
Med 3:13541361. doi:10.1038/nm1297-1354
21. Palu G, Cavaggioni A, Calvi P, Franchin E, Pizzato M, Boschetto
R, Parolin C, Chilosi M, Ferrini S, Zanusso A, Colombo F (1999)
Gene therapy of glioblastoma multiforme via combined expression of suicide and cytokine genes: a pilot study in humans. Gene
Ther 6:330337. doi:10.1038/sj.gt.3300805
22. Eck SL, Alavi JB, Alavi A, Davis A, Hackney D, Judy K,
Mollman J, Phillips PC, Wheeldon EB, Wilson JM (1996)
Treatment of advanced CNS malignancies with the recombinant
adenovirus H5.010RSVTK: a phase I trial. Hum Gene Ther
7:14651482. doi:10.1089/hum.1996.7.12-1465
23. Klatzmann D, Valery CA, Bensimon G, Marro B, Boyer O,
Mokhtari K, Diquet B, Salzmann JL, Philippon J (1998) A phase
I/II study of herpes simplex virus type 1 thymidine kinase suicide gene therapy for recurrent glioblastoma. Study Group on
Gene Therapy for Glioblastoma. Hum Gene Ther 9:25952604.
doi:10.1089/hum.1998.9.17-2595
24. Kun LE, Gajjar A, Muhlbauer M, Heideman RL, Sanford R,
Brenner M, Walter A, Langston J, Jenkins J, Facchini S (1995)
Stereotactic injection of herpes simplex thymidine kinase vector
producer cells (PA317G1Tk1SvNa.7) and intravenous ganciclovir for the treatment of progressive or recurrent primary
supratentorial pediatric malignant brain tumors. Hum Gene Ther
6:12311255. doi:10.1089/hum.1995.6.9-1231
25. Oldfield EH, Ram Z, Culver KW, Blaese RM, DeVroom HL,
Anderson WF (1993) Gene therapy for the treatment of brain
tumors using intra-tumoral transduction with the thymidine
kinase gene and intravenous ganciclovir. Hum Gene Ther 4:39
69. doi:10.1089/hum.1993.4.1-39
26. Raffel C, Culver K, Kohn D, Nelson M, Siegel S, Gillis F, Link
CJ, Villablanca JG, Anderson WF (1994) Gene therapy for the

271

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

treatment of recurrent pediatric malignant astrocytomas with in


vivo tumor transduction with the herpes simplex thymidine
kinase gene/ganciclovir system. Hum Gene Ther 5:863890. doi:
10.1089/hum.1994.5.7-863
Smith JS, Alderete B, Minn Y, Borell TJ, Perry A, Mohapatra G,
Hosek SM, Kimmel D, OFallon J, Yates A, Feuerstein BG,
Burger PC, Scheithauer BW, Jenkins RB (1999) Localization of
common deletion regions on 1p and 19q in human gliomas and
their association with histological subtype. Oncogene 18:4144
4152. doi:10.1038/sj.onc.1202759
White FV, Dehner LP, Belchis DA, Conard K, Davis MM,
Stocker JT, Zuppan CW, Biegel JA, Perlman EJ (1999) Congenital disseminated malignant rhabdoid tumor: a distinct
clinicopathologic entity demonstrating abnormalities of chromosome 22q11. Am J Surg Pathol 23:249256. doi:10.1097/00000
478-199903000-00001
Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, Peter
M, Kovar H, Joubert I, de Jong P, Rouleau G et al (1992) Gene
fusion with an ETS DNA-binding domain caused by chromosome
translocation in human tumours. Nature 359:162165. doi:
10.1038/359162a0
Maruno M, Yoshimine T, Muhammad AK, Ninomiya H, Kato A,
Hayakawa T (1999) Chromosomal aberrations detected by comparative genomic hybridization (CGH) in human astrocytic tumors.
Cancer Lett 135:6166. doi:10.1016/S0304-3835(98)00266-3
Bayani J, Zielenska M, Marrano P, Kwan Ng Y, Taylor MD, Jay
V, Rutka JT, Squire JA (2000) Molecular cytogenetic analysis of
medulloblastomas and supratentorial primitive neuroectodermal
tumors by using conventional banding, comparative genomic
hybridization, and spectral karyotyping. J Neurosurg 93:437448
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. doi:
10.1126/science.1068597
Fuller GN, Hess KR, Rhee CH, Yung WK, Sawaya RA, Bruner
JM, Zhang W (2002) Molecular classification of human diffuse
gliomas by multidimensional scaling analysis of gene expression
profiles parallels morphology-based classification, correlates with
survival, and reveals clinically-relevant novel glioma subsets.
Brain Pathol 12:108116
Li A, Walling J, Kotliarov Y, Center A, Steed ME, Ahn SJ,
Rosenblum M, Mikkelsen T, Zenklusen JC, Fine HA (2008)
Genomic changes and gene expression profiles reveal that
established glioma cell lines are poorly representative of primary
human gliomas. Mol Cancer Res 6:2130. doi:10.1158/15417786.MCR-07-0280
Tanwar MK, Gilbert MR, Holland EC (2002) Gene expression
microarray analysis reveals YKL-40 to be a potential serum
marker for malignant character in human glioma. Cancer Res
62:43644368
van den Boom J, Wolter M, Kuick R, Misek DE, Youkilis AS,
Wechsler DS, Sommer C, Reifenberger G, Hanash SM (2003)
Characterization of gene expression profiles associated with glioma progression using oligonucleotide-based microarray analysis
and real-time reverse transcription-polymerase chain reaction.
Am J Pathol 163:10331043
Mehrian Shai R, Reichardt JK, Ya-Hsuan H, Kremen TJ, Liau
LM, Cloughesy TF, Mischel PS, Nelson SF (2005) Robustness of
gene expression profiling in glioma specimen samplings and
derived cell lines. Brain Res Mol Brain Res 136:99103. doi:
10.1016/j.molbrainres.2005.01.017
Tsai MH, Cook JA, Chandramouli GV, DeGraff W, Yan H, Zhao
S, Coleman CN, Mitchell JB, Chuang EY (2007) Gene expression
profiling of breast, prostate, and glioma cells following single
versus fractionated doses of radiation. Cancer Res 67:38453852.
doi:10.1158/0008-5472.CAN-06-4250

123

272
39. Osawa H, Smith C, Ra YS, Rutka JT (2009) The role of the
membrane cytoskeleton crosslinker ezrin in medulloblastoma
cells. Neuro-oncol. doi:10.1215/15228517-2008-110
40. Tamber MS, Bansal K, Liang ML, Mainprize TG, Salhia B,
Northcott P, Taylor M, Rutka JT (2006) Current concepts in the
molecular genetics of pediatric brain tumors: implications for
emerging therapies. Childs Nerv Syst 22:13791394. doi:
10.1007/s00381-006-0187-3
41. Cowell JK, Matsui S, Wang YD, LaDuca J, Conroy J, McQuaid D,
Nowak NJ (2004) Application of bacterial artificial chromosome
array-based comparative genomic hybridization and spectral
karyotyping to the analysis of glioblastoma multiforme. Cancer
Genet Cytogenet 151:3651. doi:10.1016/j.cancergencyto.2003.
09.012
42. 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. doi:10.1158/1078-0432.CCR-05-0128
43. Rossi MR, Conroy J, McQuaid D, Nowak NJ, Rutka JT, Cowell
JK (2006) Array CGH analysis of pediatric medulloblastomas.
Genes Chromosomes Cancer 45:290303. doi:10.1002/gcc.20292
44. Taylor MD, Poppleton H, Fuller C, Su X, Liu Y, Jensen P,
Magdaleno S, Dalton J, Calabrese C, Board J, Macdonald T,
Rutka J, Guha A, Gajjar A, Curran T, Gilbertson RJ (2005)
Radial glia cells are candidate stem cells of ependymoma. Cancer
Cell 8:323335. doi:10.1016/j.ccr.2005.09.001
45. Cancer genome atlas research network (2008) Comprehensive
genomic characterization defines human glioblastoma genes and
core pathways. Nature 455:10611068. doi:10.1038/nature07385
46. Northcott P, Nakahara Y, Xiaochong W, et al. (2009) Multiple
recurrent genetic events converge on control of histone lysine
methylation in medulloblastoma. Nature Genetics. doi:10.1038/
ng.336
47. Bird A (2002) DNA methylation patterns and epigenetic memory.
Genes Dev 16:621. doi:10.1101/gad.947102
48. Bhaumik SR, Smith E, Shilatifard A (2007) Covalent modifications of histones during development and disease pathogenesis.
Nat Struct Mol Biol 14:10081016. doi:10.1038/nsmb1337
49. Gronbaek K, Hother C, Jones PA (2007) Epigenetic changes in
cancer. APMIS 115:10391059. doi:10.1111/j.1600-0463.2007.
apm_636.xml.x
50. Amatya VJ, Naumann U, Weller M, Ohgaki H (2005) TP53
promoter methylation in human gliomas. Acta Neuropathol
110:178184. doi:10.1007/s00401-005-1041-5
51. Baeza N, Weller M, Yonekawa Y, Kleihues P, Ohgaki H (2003)
PTEN methylation and expression in glioblastomas. Acta Neuropathol 106:479485. doi:10.1007/s00401-003-0748-4
52. Wiencke JK, Zheng S, Jelluma N, Tihan T, Vandenberg S,
Tamguney T, Baumber R, Parsons R, Lamborn KR, Berger MS,
Wrensch MR, Haas-Kogan DA, Stokoe D (2007) Methylation of
the PTEN promoter defines low-grade gliomas and secondary
glioblastoma. Neuro-oncol 9:271279. doi:10.1215/152285172007-003
53. Foltz G, Ryu GY, Yoon JG, Nelson T, Fahey J, Frakes A, Lee H,
Field L, Zander K, Sibenaller Z, Ryken TC, Vibhakar R, Hood L,
Madan A (2006) Genome-wide analysis of epigenetic silencing
identifies BEX1 and BEX2 as candidate tumor suppressor genes
in malignant glioma. Cancer Res 66:66656674. doi:
10.1158/0008-5472.CAN-05-4453
54. Kim TY, Zhong S, Fields CR, Kim JH, Robertson KD (2006)
Epigenomic profiling reveals novel and frequent targets of
aberrant DNA methylation-mediated silencing in malignant glioma. Cancer Res 66:74907501. doi:10.1158/0008-5472.CAN05-4552

123

J Neurooncol (2009) 92:261273


55. Lee JS, Gil JE, Kim JH, Kim TK, Jin X, Oh SY, Sohn YW, Jeon
HM, Park HJ, Park JW, Shin YJ, Chung YG, Lee JB, You S, Kim
H (2008) Brain cancer stem-like cell genesis from p53-deficient
mouse astrocytes by oncogenic Ras. Biochem Biophys Res
Commun 365:496502. doi:10.1016/j.bbrc.2007.11.005
56. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N,
Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L,
Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC,
Stupp R (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352:9971003. doi:
10.1056/NEJMoa043331
57. Martinez R, Schackert G (2007) Epigenetic aberrations in
malignant gliomas: an open door leading to better understanding
and treatment. Epigenetics 2:147150
58. Lindsey JC, Anderton JA, Lusher ME, Clifford SC (2005) Epigenetic events in medulloblastoma development. Neurosurg
Focus 19:E10. doi:10.3171/foc.2005.19.5.11
59. Kongkham P, Northcott P, Taylor MD, Smith C, Mainprize T,
Croul S, Rutka JT (2008) An genome wide screen identifies
SPINT2 as a novel tumor suppressor silenced by methylation in
medulloblastoma. Cancer Res 68:99459953. doi:10.1158/00085472.CAN-08-2169
60. Vibhakar R, Foltz G, Yoon JG, Field L, Lee H, Ryu GY, Pierson
J, Davidson B, Madan A (2007) Dickkopf-1 is an epigenetically
silenced candidate tumor suppressor gene in medulloblastoma.
Neuro-oncol 9:135144. doi:10.1215/15228517-2006-038
61. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense
complementarity to lin-14. Cell 75:843854. doi:10.1016/00928674(93)90529-Y
62. Wightman B, Ha I, Ruvkun G (1993) Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal
pattern formation in C. elegans. Cell 75:855862. doi:10.1016/
0092-8674(93)90530-4
63. Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ (2008)
miRBase: tools for microRNA genomics. Nucleic Acids Res
36:D154D158. doi:10.1093/nar/gkm952
64. Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281297. doi:10.1016/S0092-8674
(04)00045-5
65. Alvarez-Garcia I, Miska EA (2005) MicroRNA functions in
animal development and human disease. Development 132:4653
4662. doi:10.1242/dev.02073
66. Stefani G, Slack FJ (2008) Small non-coding RNAs in animal
development. Nat Rev Mol Cell Biol 9:219230. doi:10.1038/
nrm2347
67. Calin GA, Croce CM (2006) MicroRNA signatures in human
cancers. Nat Rev Cancer 6:857866. doi:10.1038/nrc1997
68. Esquela-Kerscher A, Slack FJ (2006) Oncomirs - microRNAs
with a role in cancer. Nat Rev Cancer 6:259269. doi:10.1038/
nrc1840
69. Hammond SM (2006) MicroRNAs as oncogenes. Curr Opin
Genet Dev 16:49. doi:10.1016/j.gde.2005.12.005
70. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle
J, Bartel DP, Linsley PS, Johnson JM (2005) Microarray analysis
shows that some microRNAs downregulate large numbers of target
mRNAs. Nature 433:769773. doi:10.1038/nature03315
71. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E,
Aldler H, Rattan S, Keating M, Rai K, Rassenti L, Kipps T,
Negrini M, Bullrich F, Croce CM (2002) Frequent deletions and
down-regulation of micro- RNA genes miR15 and miR16 at
13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA
99:1552415529. doi:10.1073/pnas.242606799
72. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, Croce CM

J Neurooncol (2009) 92:261273

73.

74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

(2004) Human microRNA genes are frequently located at fragile


sites and genomic regions involved in cancers. Proc Natl Acad
Sci USA 101:29993004. doi:10.1073/pnas.0307323101
Mendell JT (2008) miRiad roles for the miR-1792 cluster in
development and disease. Cell 133:217222. doi:10.1016/j.cell.
2008.04.001
Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K,
Tomida S, Yatabe Y, Kawahara K, Sekido Y, Takahashi T (2005)
A polycistronic microRNA cluster, miR-1792, is overexpressed
in human lung cancers and enhances cell proliferation. Cancer
Res 65:96289632. doi:10.1158/0008-5472.CAN-05-2352
He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D,
Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ,
Hammond SM (2005) A microRNA polycistron as a potential
human oncogene. Nature 435:828833. doi:10.1038/nature03552
Metzler M, Wilda M, Busch K, Viehmann S, Borkhardt A (2004)
High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes Chromosomes Cancer 39:
167169. doi:10.1002/gcc.10316
Chan JA, Krichevsky AM, Kosik KS (2005) MicroRNA-21 is an
antiapoptotic factor in human glioblastoma cells. Cancer Res
65:60296033. doi:10.1158/0008-5472.CAN-05-0137
Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F,
Visone R, Iorio M, Roldo C, Ferracin M, Prueitt RL, Yanaihara
N, Lanza G, Scarpa A, Vecchione A, Negrini M, Harris CC,
Croce CM (2006) A microRNA expression signature of human
solid tumors defines cancer gene targets. Proc Natl Acad Sci USA
103:22572261. doi:10.1073/pnas.0510565103
Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H,
Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y, Mitsudomi
T, Takahashi T (2004) Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened
postoperative survival. Cancer Res 64:37533756. doi:10.1158/
0008-5472.CAN-04-0637
Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R,
Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ (2005)
RAS is regulated by the let-7 microRNA family. Cell 120:635
647. doi:10.1016/j.cell.2005.01.014
Corsten MF, Miranda R, Kasmieh R, Krichevsky AM, Weissleder
R, Shah K (2007) MicroRNA-21 knockdown disrupts glioma
growth in vivo and displays synergistic cytotoxicity with neural
precursor cell delivered S-TRAIL in human gliomas. Cancer Res
67:89949000. doi:10.1158/0008-5472.CAN-07-1045
Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M,
Vandenberg SR, Ginzinger DG, James CD, Costello JF, Bergers
G, Weiss WA, Alvarez-Buylla A, Hodgson JG (2008) miR-124
and miR-137 inhibit proliferation of glioblastoma multiforme
cells and induce differentiation of brain tumor stem cells. BMC
Med 6:14. doi:10.1186/1741-7015-6-14
Pierson J, Hostager B, Fan R, Vibhakar R (2008) Regulation of
cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. J Neurooncol 90:17. doi:10.1007/s11060-008-9624-3
Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A,
Nuovo G, Raychaudhury A, Newton HB, Chiocca EA, Lawler S
(2008) Targeting of the Bmi-1 oncogene/stem cell renewal factor
by microRNA-128 inhibits glioma proliferation and self-renewal.

273

85.

86.

87.
88.

89.

90.

91.

92.

93.

94.

Cancer Res 68:91259130. doi:10.1158/0008-5472.CAN-082629


Northcott P, Fernandez-Li A, Hagah JP, Ellison DW, Grajkowska
W, Gillespie Y, Grundy R, Van Meter T, Rutka JT, Croce CM,
Kenney AM, Taylor MD (2009) The miR-17/92 polycistron is
amplified and upregulated in Sonic Hedgehog-drive medulloblastomas and induced by N-Myc in Sonic Hedgehog-treated
cerebellar neural precursors. Cancer Res (in press)
Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P,
Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R,
Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA,
Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL,
Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin
R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE,
Kinzler KW (2008) An integrated genomic analysis of human
glioblastoma multiforme. Science 321:18071812. doi:10.1126/
science.1164382
Rusk N, Kiermer V (2008) Primer: Sequencingthe next generation. Nat Methods 5:15. doi:10.1038/nmeth1155
Hodges E, Xuan Z, Balija V, Kramer M, Molla MN, Smith SW,
Middle CM, Rodesch MJ, Albert TJ, Hannon GJ, McCombie WR
(2007) Genome-wide in situ exon capture for selective resequencing. Nat Genet 39:15221527. doi:10.1038/ng.2007.42
Campbell PJ, Stephens PJ, Pleasance ED, OMeara S, Li H, Santarius T, Stebbings LA, Leroy C, Edkins S, Hardy C, Teague JW,
Menzies A, Goodhead I, Turner DJ, Clee CM, Quail MA, Cox A,
Brown C, Durbin R, Hurles ME, Edwards PA, Bignell GR, Stratton
MR, Futreal PA (2008) Identification of somatically acquired
rearrangements in cancer using genome-wide massively parallel
paired-end sequencing. Nat Genet 40:722729. doi:10.1038/ng.128
Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E,
Orlov YL, Zhang W, Jiang J, Loh YH, Yeo HC, Yeo ZX, Narang
V, Govindarajan KR, Leong B, Shahab A, Ruan Y, Bourque G,
Sung WK, Clarke ND, Wei CL, Ng HH (2008) Integration of
external signaling pathways with the core transcriptional network
in embryonic stem cells. Cell 133:11061117. doi:10.1016/j.cell.
2008.04.043
Robertson AG, Bilenky M, Tam A, Zhao Y, Zeng T, Thiessen N,
Cezard T, Fejes AP, Wederell ED, Cullum R, Euskirchen G,
Krzywinski M, Birol I, Snyder M, Hoodless PA, Hirst M, Marra
MA, Jones SJ (2008) Genome-wide relationship between histone
H3 lysine 4 mono- and tri-methylation and transcription factor
binding. Genome Res 18:19061917
Robertson G, Hirst M, Bainbridge M, Bilenky M, Zhao Y, Zeng
T, Euskirchen G, Bernier B, Varhol R, Delaney A, Thiessen N,
Griffith OL, He A, Marra M, Snyder M, Jones S (2007) Genomewide profiles of STAT1 DNA association using chromatin
immunoprecipitation and massively parallel sequencing. Nat
Methods 4:651657. doi:10.1038/nmeth1068
Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B
(2008) Mapping and quantifying mammalian transcriptomes by
RNA-Seq. Nat Methods 5:621628. doi:10.1038/nmeth.1226
Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ (2008) Deep
surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet 40:1413
1415. doi:10.1038/ng.259

123

You might also like