Ningaraj et al.indd


Drug Target Insights 2007: 2 197–207 197

REVIEW

Correspondence: Ningaraj, N.S., Department of Pediatric Neurooncology and Molecular Pharmacology,
Hoskins Center, Curtis and Elizabeth Anderson Cancer Institute, Memorial Health University Medical Center, 
Mercer University Medical School, 4700 Waters Avenue, Savannah, GA 31404, U.S.A.Tel: +1 9123500958;
Fax: +1 9123501269; Email: NingaNa1@memorialhealth.com 
Please note that this article may not be used for commercial purposes. For further information please refer to the copyright 
statement at http://www.la-press.com/copyright.htm

Targeted Brain Tumor Treatment-Current Perspectives
Ningaraj N.S1, Salimath B.P2, Sankpal U.T1, Perera R1 and Vats T1
1Department of Pediatric Neurooncology and Molecular Pharmacology, 
Hoskins Center, Curtis and Elizabeth Anderson Cancer Institute, Memorial Health University Medical 
Center, Mercer University Medical School, 4700 Waters Avenue, Savannah, GA 31404, U.S.A.
2Department of Biotechnology, University of Mysore, Mysore 570006, Karnataka, India.

Abstract: Brain tumor is associated with poor prognosis. The treatment option is severely limited for a patient with brain 
tumor, despite great advances in understanding the etiology and molecular biology of brain tumors that have lead to break-
throughs in developing pharmaceutical strategies, and ongoing NCI/Pharma-sponsored clinical trials. We reviewed the 
literature on molecular targeted agents in preclinical and clinical studies in brain tumor for the past decade, and observed 
that the molecular targeting in brain tumors is complex. This is because no single gene or protein can be affected by single 
molecular agent, requiring the use of combination molecular therapy with cytotoxic agents. In this review, we briefl y discuss 
the potential molecular targets, and the challenges of targeted brain tumor treatment. For example, glial tumors are associ-
ated with over-expression of calcium-dependent potassium (KCa) channels, and high grade glioma express specifi c KCa 
channel gene (gBK) splice variants, and mutant epidermal growth factor receptors (EGFRvIII). These specifi c genes are 
promising targets for molecular targeted treatment in brain tumors. In addition, drugs like Avastin and Gleevec target the 
molecular targets such as vascular endothelial cell growth factor receptor, platelet-derived growth factor receptors, and 
BRC-ABL/Akt. Recent discovery of non-coding RNA, specifi cally microRNAs could be used as potential targeted drugs. 
Finally, we discuss the role of anti-cancer drug delivery to brain tumors by breaching the blood-brain tumor barrier. This 
non-invasive strategy is particularly useful as novel molecules and humanized monoclonal antibodies that target receptor 
tyrosine kinase receptors are rapidly being developed.

Abbreviations: BBB: blood-brain barrier; BTB: blood-tumor barrier; KCa: calcium-dependent potassium channels; NS-1619/
NS 004: 1,3-dihydro-1-5-(trifl uoromethyl)-2H benzimidazol-2-one; HBMVEC: human brain microvascular endothelial cells; 
FACS: fl uorescence activated cell sorting; PDGFR: platelet-derived growth factor receptor; RTKIs: receptor tyrosine kinase 
inhibitors; EGFR: epidermal growth factor receptor; EGFRvIII: variant III of the human EGFR; gBK channel: glioma spe-
cifi c spice variant of KCa channel gene; KATP:  ATP sensitive potassium channels; Minoxidil sulfate (MS: KATP channel agonist); 
Trastuzumab (Herceptin, Her-2 inhibitor, Genentech Inc.).

Keywords: Brain tumor, BBB, drug delivery, therapeutic targets in brain tumors

Introduction

Brain tumor
Nearly 20,000 new primary brain tumors and about 200,000 metastatic brain tumor cases are reported 
each year in the U.S.A. (Levin, 2007). The overall survival of these patients is dismal and the majority of 
survivors suffer disabling toxicities from their treatments. Standard treatment for brain tumors includes 
combination of surgery, radiation therapy, and chemotherapy. Brain tumor poses unique challenges due 
to its distinct biology, genetics, treatment response, and survival. Despite extensive characterization of the 
brain tumor pathways, molecularly targeted approach is not available to brain tumor patients. Future 
research in brain tumors needs to focus on strategies for improving drug delivery, disruption of 
blood-brain-barrier (BBB), and molecular profi ling of tumors. In addition, careful studies are needed to 
delineate pathways that aid and abate brain tumor progression. Identifi cation of potential markers (genes 
and proteins) for targeted therapy will defi nitely help the clinicians to design the treatment accordingly. 
Usually, after surgical treatment, brain tumor recurs, severely shortening life expectancy (Friedman, Kerby 
and Calvert, 2000). Conventional treatments using radiation and intravenous chemotherapy are not sucessful 



198

Ningaraj et al

Drug Target Insights 2007: 2

because the cancer cells develop resistance to treat-
ment. Anti-cancer drugs fail to penetrate the BBB 
in suffi cient quantities (Pardridge, 2001), allowing 
cancer cells to develop resistance to these agents. 
Therefore, understanding the biochemical regulation 
of the BBB (Fig. 1) in normal and tumor-invaded 
brain is of great importance to develop therapeutics 
that breach or circumvent BBB and directly target 
brain tumor cells (Ningaraj, 2006). The focus is now 
on the targeted cancer therapies (Butowski and 
Chang, 2005) that complement conventional treat-
ments and reduce the drug resistance in cancer cells 
and the toxicity in normal brain (Newton, 2003). 
Novel cancer therapies include anti-angiogenic 
agents, immunotherapy, bacterial agents, viral 
oncolysis, cyclin-dependent kinases and receptor 
tyrosine kinase inhibitors (RTKIs), anti-sense 
agents, gene therapy, microRNA (miRNA), and 
combinations of various methods (Butowski and 
Chang, 2005). 

Chemotherapy
Chemotherapy is a form of targeted therapy where 
cytotoxic drugs act on multiplying tumor cells. The 
drugs can also be used as sensitizers to augment the 
effects of radiation therapy. Chemotherapeutic 
drugs can be delivered directly to brain tumors 
through a polymer wafer implant such as a biode-
gradable wafer soaked with BCNU (Carmustine). 
Besides BCNU, several other chemotherapy drugs 
are used to treat brain tumors, which are adminis-
tered by various routes. The chemotherapeutic 
drugs taken orally include Temozolomide (TMZ, 
Temodar), procarbazine (Matulane), and lomustine 

(CCNU). The intravenously administered drugs 
include vincristine (Oncovin or Vincasar PFS), 
cisplatin (Platinol), carmustine (BCNU, BiCNU), 
Carboplatin (Paraplatin), while Methotrexate 
(Rheumatrex or Trexall) may be taken orally, by 
injection, or intrathecally. Treating brain tumors 
with chemotherapy can be diffi cult because the 
brain is protected by BBB, which keeps out harmful 
substances such as bacteria and chemotherapeutic 
drugs. Among many cytotoxic agents in the clini-
cian’s arsenal, Temozolomide (TMZ) has shown 
some promise in treatment of low grade gliomas 
(Friedman, Kerby and Calvert, 2000), however, the 
effect on patient survival was modest (Balana et al. 
2004). The problem is that glioblastoma multiforme 
(GBM) exhibits varying responses to TMZ (Hirose, 
Berger and Pieper, 2001), and in some cases 
gliomas have increased O6-methyl guanine methyl 
transferase (MGMT) activity, which results in 
complete resistance to TMZ (Bocangel et al. 2002). 
The clinical utility of TMZ against all types of brain 
tumors remains limited due to its BTB penetration 
(some authors claim TMZ metabolite (MTIC) 
concentration in CSF to be as high as 30%), which 
demand repeated high doses to achieve in vivo 
therapeutically effective concentrations in brain 
tumors (Yung et al. 1999), and different phenotypes 
and genotypes that render some form of resistance 
against TMZ (Kanzawa et al. 2003). Most 
importantly, an extensive literature search and 
preliminary work on BBB/BTB penetration of TMZ 
did not convince us that suffi cient amount of drug 
penetrates the BBB or BTB to elicit anti-tumor 
effect. To circumvent the penetration problem, 
chemotherapy drugs can be delivered by 

Figure 1. A schematic representation of normal and abnormal blood-brain barriers. We reason that genes in brain cancers/vasculature are 
distinct from normal brain/vasculature. They are attractive targets for the design of therapies that can penetrate the BTB and selectively kill brain 
cancer cells. We are studying the genes that direct the formation of the normal and abnormal (cancer) human brain vasculature, and with this 
knowledge develop new treatment strategies for brain tumor patients. EC: endothelial cells; TJ: tight junction proteins; N: nucleus. 



199

Targeted Brain Tumor Treatment

Drug Target Insights 2007: 2 

intratumoral route or by drug impregnated wafers 
to attain higher concentration of drugs in the tumor 
cells, but the procedures are highly invasive.

Targeted brain tumor treatment
The human genome project has raised the expecta-
tion of the development of novel therapies for brain 
tumor because the conventional treatment strate-
gies have not yielded any significant clinical 
outcome. Brain tumor treatment differs according 
to the grade and location of the tumor. Hence, 
combination of surgery, chemotherapy, and radio-
therapy can be used in treating brain tumor patients 
(Stupp et al. 2005). Most promising anti-cancer 
drugs for pediatric and adult patients that are effec-
tive against cancers outside the brain have failed 
against brain tumors in clinical trails, in part, due 
to poor penetration across the BBB. For instance, 
aberrant expression of src family kinase (LCK) 
(Fabian et al. 2005) or mutation of c-KIT are 
involved in the pathogenesis of many cancers. 
Studies using imatinib mesylate (STI 571, Gleevec, 
Novartis, U.S.A.), an inhibitor of the tyrosine 
kinases BRC-ABL, c-KIT, and PDGFR, have 
shown signifi cant response in patients with chronic 
myelogenous leukemia (CML) and gastrointestinal 
stromal tumor (GIST). Clinical trials were recently 
conducted to test the effi cacy of Gleevec in brain 
tumors (Reardon et al. 2005; Wen et al. 2006; 
Pollack et al. 2007). Gleevec is an effective agent 
that targets specifi c gene/protein in cancer cells 
without harming normal cells and tissues. Drugs 
like Gleevec and Temozolomide attack abnormal 
chemical signals or molecules inside the cells or 
on the surface of the cells that have enabled brain 
tumor cells to escape the normal growth controls. 
Therefore, combating many forms of cancer will 
probably require a variety of targeted drugs used 
in combination, as cancer involves different types 
of dysfunctional genes and no single or two drugs 
will be sufficient. Some cancers, particularly 
primary and metastatic brain tumors of the breast 
and lung are diffi cult to treat because they are 
caused by multiple signaling pathways that are 
running amok, rather than just one, as observed in 
CML and GIST (Butowski and Chang, 2005). 
Gleevec may potentially target the above mentioned 
oncogenes in brain tumor (Holdhoff et al. 2005) 
provided it penetrates the BBB (Leis et al. 2004). 
Careful molecular studies would identify the stem 
cell factor/c-kit pathways in pediatric brain tumors, 
which might be the target of Gleevec. Characterizing 

the genetic and proteomic events that play a role 
in the biology of these tumors may allow molecular 
sub-typing which could lead to the development 
of novel therapeutic strategies, including treatment 
with Gleevec or with potassium channel modula-
tors targeting tumor and tumor blood vessel endo-
thelial cells (Ningaraj, 2006).

Targeting brain tumors 
Targeting tumor and tumor blood vessel-specific 
marker(s) is a good strategy to control tumor 
growth (Robinson et al. 2003). It is, however, 
critical to study whether tumor-specific drug 
delivery has the potential to minimize toxicity 
to normal tissues, and to improve the bioavail-
ability of cytotoxic agents to neoplasms. 
Existing site-specific drug delivery systems 
include delivery to endothelial receptor αvβ3, 
and tumor specific antigens. Antibody conjuga-
tion to cytotoxic agents has shown promise in 
achieving the goal of tumor-targeted cytotox-
icity. This approach may be limited by the small 
subsets of tumors that can be targeted by these 
antibodies and by poor biodistribution of these 
a n t i b o d i e s  i n t o  s o l i d  t u m o r s .  A l t e r n a t i v e 
approaches to target all neoplasms exploit 
differences in human tumor blood vessel char-
acteristics when compared to normal brain blood 
vessels (Black and Ningaraj, 2004; Ningaraj 
et al. 2002; Ningaraj, Rao and Black, 2003a). 

Epidermal growth factor receptor (EGFR) is 
often amplified and mutated in human gliomas, 
but the expression is low or undetectable in 
normal brain. Recently, EGFR’s mutant isoform, 
variant III of the human EGFR (EGFRvIII), is 
under intensive investigation as potential 
molecular target for the specific delivery of the 
diagnostic and the therapeutic agents to brain 
tumors (Yang et al. 2005). The therapeutic 
monoclonal antibodies (MAb) targeting growth 
factor pathways are being developed. The 
purpose of antibody treatment of cancer is to 
induce the direct or indirect destruction of 
cancer cells, either by specifically targeting the 
tumor or the tumor vasculature (Butowski and 
Chang, 2005). Examples of therapeutic anti-
bodies which are effective in treating cancer 
includes the humanized IgG antibody Herceptin 
for the treatment of breast cancer, Cetuximab, 
ABX-EGF, EMD 720000 and h-R3 directed at 
extracellular receptor domain that inhibits the 
ligand-receptor interactions. Other antibodies 



200

Ningaraj et al

Drug Target Insights 2007: 2

like Y10 and MAb806, which are directed 
towards the extracellular portion of EGFRvIII 
in gliomas have also shown some activity in 
clinical trials (Rich and Bigner, 2004). Suramin 
(polysulfonated napthylurea), which acts by 
interfering with the binding of several growth 
factors-including EGF, platelet derived growth 
factor (PDGF), and insulin growth factor (IGF1) 
with their putative receptors, is being tested in 
clinical trails. These MAbs, however, have poor 
penetration into brain tumors, which results in 
recurrence in brain tumor patients. 

Kinase inhibitors
Kinase inhibitors show great promise as a new 
class of therapeutics to control gliomas. The 
specifi city of RTKIs, including those that are in 
clinical use or in development widely varies, and 
is not strongly correlated with chemical structure 
of the identity of the intended target. Many novel 
interactions were recently identifi ed (Fabian et al. 
2005). EGFR and PDGFR are abnormal genes 
identifi ed in gliomas (Rich and Bigner, 2004), 
whose expression is linked to an increased rate of 
tumor cell proliferation, resistance to chemo-
therapy, invasion, and apoptosis, and hence 
decreased survival in patients with malignant 
gliomas. PDGF ligands bind to PDGFRs to induce 
phosphorylation and activation of downstream 
signaling pathways such as RAS, MAPK, and Akt. 
Therefore, therapies using Gleevec, Suramin, and 
MAbs are directed at PDGFR to control glioma 
growth. PDGFR inhibitors may also provide addi-
tional benefit by blocking pericytes-assisted 
angiogenesis (Bergers et al. 2003). Clinical trials 
with EGFR and PDGFR inhibitors have shown 
promise for glioma therapy, although their ability 
to penetrate BBB in suffi cient amounts is largely 
unknown. We transiently opened the BTB with 
KCa and ATP-sensitive potassium (KATP) channel 
agonists (Black and Ningaraj, 2004; Ningaraj et al. 
2002; Ningaraj, Rao and Black, 2003a,b; Rao and 
Ningaraj, 2001) to increase the delivery of Gleevec 
and Herceptin to human glioma xenografts grown 
in murine brains.

KCa channels in gliomas
Membrane ion channels are essential for cell 
proliferation and appears to play a role in the 
development of cancer (Ningaraj, 2006). The 

KCa channels are highly expressed in gliomas 
(Weaver, Liu and Sontheimer, 2004) supporting 
the hypothesis that these channels play an 
important role in brain tumor growth and 
possibly the progression of low grade anaplastic 
astrocytomas (grade II) to a deadly high grade 
GBM (WHO grade IV). In addition, studies have 
shown that modulation of the biological function 
of KCa channels with specific inhibitors atten-
uate glioma growth (Rao and Ningaraj, 2001). 
Another study showed that the activation of 
intermediate KC a channels with its opener 
caused down-regulation of these channels and 
attenuated the non-excitable cell growth and its 
proliferation (Kraft et al. 2003). We showed that 
chronic activation of KCa channels with its 
specific openers NS-1619 and NS-004 elicited 
apoptosis in vitro and in vivo (Rao and Ningaraj, 
2001). However, the role of KCa channels in 
progression from a treatable low grade to an 
untreatable high grade glioma in pediatric as 
well as in adult patients is not fully understood. 
Recently, glioma KCa/BK channels (gBK) splice 
variant of the KCNMA1 gene was characterized 
by enhanced sensitivity to intracellular calcium 
levels (Weaver, Bomben and Sontheimer, 2006). 
The study also showed that the expression of 
functional gBK channels appears to be regulated 
in a growth-factor-dependent manner. It is well 
established that EGF activates EGFR. Several 
molecular agents targeting EGFR are under-
going clinical trails as potential therapies in 
neurooncology (Rich and Bigner, 2004). For 
example, ZD1839 (Iressa) an orally active, 
selective EGFR-tyrosine kinase inhibitor has 
anti-tumor activity against malignant human 
cancer cell lines (31). Glioma cells also show 
up-regulation and constitutive activation of Her-
2 neu, and its expression which correlates posi-
tively with aggressive malignancy (Mellinghoff 
et al. 2005). A correlation has been demonstrated 
for the expression of gBK/KCa channels and 
Her-2 neu, which implies gBK/KCa channels as 
a downstream target for Her-2 neu signaling 
(Olsen et al. 2004). How KCa channel modulates 
EGFR tyrosine kinase or vice versa is poorly 
understood. It appears to occur via changes in 
intracellular calcium levels without change in 
channel expression or phosphorylation (Weaver, 
Bomben and Sontheimer, 2006). In a transgenic 
glioma mouse model, a loss of EGFR overex-
pression was observed by EGFRvIII introduction 



201

Targeted Brain Tumor Treatment

Drug Target Insights 2007: 2 

(Gullick, 2001). This model of high grade 
glioma is useful in evaluating targeted molecular 
therapies in brain tumor.

Targeting angiogenesis in brain 
tumors
Angiogenesis plays a crucial role in malignant 
primary brain tumor growth. Several preclinical 
and clinical studies have confirmed that the 
vascular endothelial cell growth factor (VEGF) 
and the bFGF bind to their receptors to promote 
glioma growth. VEGFR is expressed in human 
high grade glioma but not found in normal brain. 
Increased concentration of angiogenic factors and 
their receptors is correlated with tumor vascula-
ture and malignant human gliomas. Furthermore, 
it is shown that the endogenous inhibitor of angio-
genesis, thrombospondin-1 (TSP-1) is produced 
by normal brain and low grade gliomas, but is 
completely absent in high grade gliomas. The 
GBM is among the most “endothelial rich” brain 
tumors studied. In children with brain tumors, 
microvascular density correlates with tumor 
recurrence, and patient mortality. As tumor vascu-
larity is highly correlated with disease outcome 
in neuroblastoma, novel therapeutic that targets 
the vascular endothelium is a suitable clinical trial 
target candidate. The molecules like VEGF, bFGF, 
PDGF as well as endothelial integrins are linked 
to advanced malignancy, which provided the 
rationale for developing anti-angiogenic therapies 
in brain tumors. The potential of anti-angiogenic 
therapy in human brain tumors is demonstrated 
in experimental brain tumor models. A wide range 
of anti-angiogenic agents such as endogenous 
angiogenesis inhibitors, synthetic angiogenic 
inhibitors, antibodies, and anti-angiogenic gene 
therapy are investigated with radiation therapy. 
Anti-angiogenic drugs have low potential for 
toxicity and resistance because they specifi cally 
target endothelial cells. The potential of anti-
angiogenic agents to augment the anti-tumor 
activity of standard cytotoxic chemotherapeutic 
agents is being investigated (Bernsen and van der 
Kogel, 1999; Reijneveld, Voest and Taphoorn, 
2000; Takano et al. 2004).

The evidence for glioma anti-angiogenesis 
therapy, with or without chemotherapy has been 
described in several preclinical animal models. 
The anti-angiogenic function of TSP-1 is known 
for a long time. The TSP-1 transfected glioma 

cells lacked VEGF expression ability, which 
supports the rationale for using VEGF and bFGF 
antibodies in clinical trails. Anti-angiogenic 
d r u g ,  T h a l i d o m i d e  e x h i b i t s  s y n e r g i s t i c 
anti-glioma activity when combined with DNA 
alkylating agent Temozolomide, and increased 
median survival from 63 weeks to 103 weeks 
compared to Thalidomide only group (Baumann 
et al. 2004). While evaluating anti-angiogenic 
drugs for clinical development, it is important 
to analyze if such drugs penetrate the BBB, and 
survive p-glycoprotein-mediated drug efflux 
system. At present, there is a great deal of interest 
in combination therapy using conventional cyto-
toxic therapy with chemotherapeutics and radio-
therapy. Anti-angiogenic agents like TNP-470, 
angiostatin, DC 101, SU5416, anti-VEGF and 
VEGF-R antibodies and VEGF monoclonal 
antibody A4.6.1, tyrosine kinase inhibitors, 
COX-2 inhibitors, and anti-EGFR inhibitors are 
used in combination with radiation. The synthetic 
fumagillin analogue, TNP-470 was shown to 
interfere with angiogenesis through inhibition of 
endothelial cell proliferation and migration in 
murine and human neuroblastoma xenograft 
model. Now it is being evaluated in phase 
I/II clinical trials. In brain tumor models, TNP-
470 and Minocycline together increased 9L 
glioma sensitivity to BCNU and andriamycin 
(Shusterman et al. 2001), while Lund, Bastholm 
and Kristjansen, (2000) found that TNP-470 
increased radiation sensitivity of human U87 
glioblastoma xenografts. A phase II study with 
anti-angiogenic monoclonal antibody bevaci-
zumab (Avastin) and anti-cytokine Irinotecan in 
brain tumor patients is also being conducted 
(NCT00381797). Endogenous inhibitors of 
angiogenesis such as angiostatin, endostatin, 
PEX, pigment epithelial-derived factor, and 
thrombospondin (TSP-1&2) are shown to be 
effi cacious. They exert their effects through 
multiple mechanisms, including induction of 
apoptosis of micro vascular endothelial cells, 
inhibition of proliferation of endothelial cells, 
inhibition of function, and regulation of proan-
giogenic molecules.  These endogenous inhibi-
tors offer a novel treatment option because they 
are unlikely to trigger a host immune response. 
Angiostatin, a proteolytic fragment of plasmin-
ogen inhibits angiogenesis and attenuate the 
growth of primary and metastatic tumors. Angi-
ostatin was effectively used in combination with 



202

Ningaraj et al

Drug Target Insights 2007: 2

fractional radiation therapy in human glioma 
models (Mauceri et al. 1998; Rege, Fears and 
Gladson, 2005). Recently, gene therapy has hit 
a snag, but offers a promising alternate treatment 
strategy.

Brain tumors are attractive for gene therapy 
because the brain is an immunologically privileged 
organ, and the BBB provides a natural immuno-
logical barrier. Mice when treated with a retrovirus 
encoding a dominant negative mutant of the VEGF 
receptor Flk-1 resulted in reduced tumor growth 
and decreased blood vessel density. Recombinant 
adeno-associated virus (AAV) vector with the 
angiostatin gene was used to reduce tumor growth 
and angiogenesis in a C6 glioma model. Anti-
angiogenic therapy using Semliki Forest Virus 
(SFV) carrying endostatin gene significantly 
reduced the tumor growth in animals. Therefore, 
gene therapy with endostatin delivered via SFV 
may be a viable treatment strategy for brain tumors 
(Ma et al. 2002; Yamanaka et al. 2003). Although, 
the gene therapy in general is in its infancy, it 
provides an alternate strategy to treat hard-to treat 
brain tumors. 

Epigenetic genes as brain tumor 
targets 
Epigenetic events are genetic modifi cations (DNA 
methylation and covalent histone modifi cations) that 
are heritable through cell division, which affect gene 
expression without causing changes to the DNA 
coding sequence. Cancer cells exhibit global 
hypomethylation of the genome accompanied by 
region-specifi c hypermethylation events. The hypo-
methylation mainly occurs in the repetitive sequences 
leading to genomic instability and tumor formation. 
Aberrant hypermethylation occurs at CpG islands 
found in the promoter region of genes, which is 
usually associated with the transcriptional silencing 
of that gene (Baylin et al. 2001). DNA methylation 
changes (Palanichamy, Erkkinen and Chakravarti, 
2006), particularly CpG island hypermethylation is 
frequent, early, and common event (as common as 
mutations) in many types of cancers leading to the 
inactivation of tumor suppressor genes. 

Several genetic changes have been identifi ed in 
AAs and GBMs involving heterozygous deletion 
of 19q13, inactivation/deletion of tumor suppressor 
genes namely p16INK4A (Hegi et al. 1997), 
p14ARF (Ichimura et al. 2000), RB1 (Ichimura 
et al. 1996), PTEN and p53 gene (Mashiyama et al. 

1991) and amplifi cation of EGFR gene (Libermann 
et al. 1985). Epigenetic research in glioma patho-
genesis revealed several epigenetic genes silenced 
by promoter CpG island hypermethylation, 
such as, cell cycle regulatory proteins RB1 
(Nakamura et al. 1996), p16INK4A (Costello et al. 
1996; Fueyo et al. 1996), myelin related gene 
EMP3 (Alaminos et al. 2005), and matrix metal-
loproteinases inhibitor TIMP3 (Bachman et al. 
1999). Comprehensive whole-genome microarray 
studies using inhibitors of epigenetic modifi cation 
have identified several genes including CST6 
(putative metastatic suppressor), BIK (apoptosis 
inducer), TSPYL5 (unknown function), BEX1, and 
BEX2 (uncharacterized function) as putative tumor 
suppressors that are frequently methylated in 
primary gliomas (Kim et al. 2006; Foltz et al. 
2006). Another genome-wide study using restric-
tion landmark genomic scanning has identifi ed as 
many as 1500 CpG islands to be aberrantly meth-
ylated in low grade gliomas (Costello et al. 2000). 
These studies have highlighted a role for DNA 
methylation in gliomagenesis. To date very few 
genetic assays are available to accurately provide 
information regarding patient prognosis or 
response to therapy. It has been hypothesized that 
aberrant DNA methylation plays a key role in 
tumor initiation. Therefore identifying such 
modifi cations helps in early detection of cancer, 
and might also provide information regarding the 
mechanisms that control glioma progression 
(Costello, 2003). In addition to being a diagnostic 
marker, DNA methylation can also serve as an 
useful prognostic marker as shown by the 
methylation of the DNA repair gene, MGMT, in 
gliomas. Epigenetic silencing of the gene (involved 
in the repair of DNA damaged by alkylating agents) 
is associated with the increased survival in patients 
treated with alkylating drug Temozolomide 
(Esteller et al. 2000; Komine et al. 2003). Current 
laboratory studies are aimed at discovering novel 
methylation markers in tumor tissue as well as in 
the patient’s body fl uids (Belinsky et al. 2006; 
Cairns et al. 2001). Since the primary DNA 
sequence of epigenetically modifi ed genes remains 
intact, it is possible to reactivate genes using 
inhibitors of DNA methylation or histone modifi -
cations (Daskalakis et al. 2002; Plumb et al. 2000). 
Clinical trials using DNA methylation and histone 
deacetylase inhibitors, which reactivate silenced 
genes in cancers, are in various development 
stages. The DNA methyltransferase inhibitors, 



203

Targeted Brain Tumor Treatment

Drug Target Insights 2007: 2 

5-azacytidine (Vidaza) and 5-aza-2′-deoxycytidine 
(Decitabine), are used with reasonable success in 
the treatment of hematologic malignancies 
(Lubbert, 2000), but have limited success in solid 
tumors. Combination of HDAC inhibitors with 
DNA methyltransferase inhibitors appear to syner-
gistically induce the expression of silenced genes 
(Cameron et al. 1999). However, these drugs have 
drawbacks such as extreme instability, serious side 
effects, and sometimes these drugs at high doses 
may promote malignant transformation. Alterna-
tive approaches include the use of siRNA targeted 
against the DNA methyltransferase enzyme (Goffi n 
and Eisenhauer, 2002) and developing stable small 
molecule inhibitors that can overcome the BBB.

Small interfering RNA (siRNA) 
to target brain tumor gene(s) 
The siRNA directs the targeted destruction of 
mRNA encoding a specific protein, in a process 
known as RNA interference (RNAi). This 
process stops translation of the targeted mRNA 
into protein, effectively silencing the gene. 
RNAi is a recent discovery, identified in 
mammalian cells in 2001, but it has rapidly 
advanced into practical technique, and is being 
used increasingly to investigate mammalian 
gene function. Tools are available to induce 
RNAi in cell lines, intact tissue preparations, 
and even in in vivo. Depending on the method 
used, loss of gene expression may be transient 
or sustained, enabling a wide range of functions 
to be investigated. The RNAi is a powerful 
technique that can be used to produce targeted 
knockout of genes in mammalian cells (Gurney 
and Hunter, 2005). Its applications potentially 
include identification of protein function in 
health and disease, identification of novel genes, 
and drug target validation. Effective RNAi 
requires an appropriate siRNA sequence to be 
designed and an efficient method for delivering 
the siRNA to the cells of interest. Since not all 
potential siRNA sequences are effective, it is 
important to verify the loss of gene expression 
by measuring the level of protein remaining. 
Limitations for delivering siRNA are one of the 
main obstacles to produce efficient RNAi, espe-
cially in intact tissue preparations. A successful 
in vitro method for targeted RNAi against the 
TASK-1 potassium channel gene (Gurney and 
H u n t e r,  2 0 0 5 )  w a s  d e s c r i b e d .  I n c r e a s i n g 

evidence show that microRNA (miRNA) repre-
sent a new class of genes involved in oncogen-
esis (Ciafre et al. 2005).

miRNAs as druggable targets 
The miRNAs are non-coding, double stranded 
RNA molecules with an average size of 22 bp, and 
serve as posttranscriptional regulators of gene 
expression in higher eukaryotes. The miRNAs play 
an important role in development and other cellular 
processes by hybridizing to complementary target 
mRNA transcripts and destabilizing the latter by 
preventing their translation (Ambros, 2003; Bartel 
and Bartel, 2003; Bartel, 2004). Although a few 
hundred miRNAs have been discovered in a variety 
of organisms, little is known about their cellular 
functions. They have been implicated, among 
others, in regulation of developmental timing and 
pattern formation, restriction of differentiation 
potential, regulation of insulin secretion, resistance 
to viral infection, and in genomic rearrangements 
associated with carcinogenesis or other genetic 
disorders, such as the fragile X syndrome. Recent 
evidence suggests that the number of unique 
miRNA genes in human ranges from 1000 to 
20,000. It is estimated that 20%–30% of all human 
mRNA genes are miRNA targets, and hence special 
attention has been given to miRNAs as candidate 
drug targets in brain tumor.

Several recent reviews and research articles 
have illustrated the involvement of miRNAs in 
cancer (Calin and Croce, 2006a, 2006b; Jannot 
and Simard, 2006; Kent and Mendell, 2006;  
Jovanovic and Hengartner, 2006; Dalmay and 
Edwards, 2006; Hutvagner, 2006; Osada and 
Takahashi, 2006; Zhang and Coukos, 2006). 
Therefore, we will restrict this section to the 
general concepts. In a recent study the miRNA 
expression levels in GBM was investigated 
(Ciafre et al. 2005; Chan, Krichevsky and Kosik, 
2005). The analysis of both glioblastoma tissues 
and glioblastoma cell lines showed a signifi cantly 
altered miRNA expression. The most interesting 
miRNA is miR-21, which is signifi cantly up-
regulated in glioblastoma. In another study, 
knockdown of miRNA-21 in cultured glioblas-
toma cells triggers activation of caspases that 
leads to increased apoptotic cell death (Chan, 
Krichevsky and Kosik, 2005). These data suggest 
that aberrantly expressed miR-21 may contribute 
to the malignant phenotype by blocking expression 



204

Ningaraj et al

Drug Target Insights 2007: 2

of critical apoptosis-related genes. A set of 
brain-enriched miRNAs, miR-128, miR-181a, 
miR-181b, and miR-181c, are down-regulated in 
glioblastoma is also discovered (Ciafre et al. 2005; 
O’Driscoll, 2006). One of the early works that 
demonstrates miRNAs as potential candidate drug 
target was performed by obstructing the adipocyte 
differentiation process in human primary adipocytes 
(Esau et al. 2004). Major hurdles are expected before 
a miRNA-based drug is successfully developed 
against cancer. In fairness this is only the beginning 
of the impact of the discovery of miRNA on under-
standing the brain tumor etiology, and developing 
cancer treatment strategies.

Anti-cancer drug delivery to brain 
tumor 
Drug delivery in the treatment of brain tumors is 
a crucial consideration in the development of anti-
cancer agent because the delivery of all substances 
into the brain is tightly regulated by BBB. Brain 
tumor cells diffuse into the normal brain and are 
protected by intact BBB (Rich and Bigner, 2004), 

where anti-cancer drug delivery is very critical. 
We showed that improved drug delivery in human 
glioma xenograft models (Ningaraj, 2006) has the 
potential to be extrapolated to patients with brain 
tumors for better control of the disease. In this 
direction, our laboratory is developing methods 
for high-throughput screening of RTKIs for selec-
tive delivery to brain tumors, simultaneously 
monitor dosing, delivery, and pharmacological 
effi cacy of RTK inhibitors in animal brain tumor 
models. The challenges and opportunities of the 
biochemical modulation of BBB for selective drug 
delivery to brain tumor was reviewed recently 
(Ningaraj, 2006). We showed that intravenously 
administered, potassium channel agonists increase 
TMZ (Fig. 2) and Her-2 MAb (Herceptin) (Ning-
araj, Rao and Black, 2003b) delivery across the 
BTB to elicit anti-tumor activity and increase 
survival in nude mice with intracranially implanted 
human glial tumor. Our study suggested that the 
BTB allows a small amount of TMZ into brain 
tumors. Potassium channel agonist-mediated 
biochemical modulation signifi cantly increased 
BTB permeability allowing greater amounts of 

Figure 2. Quantitative increases in BTB permeability. A signifi cant increase in the mean Ki for [
14C]-Temozolomide (TMZ) after i.v. infusion 

of 100 µg/kg/min for 15 min of NS-1619 and MS compared to a vehicle-treated group was observed. The increase in [14C]-TMZ uptake in 
tumor center was signifi cant although a slight increase in uptake of the radiotracer was observed in the brain tissue-surrounding tumor. No 
[14C]-TMZ uptake in contralateral normal brain, which served as internal control, was observed in all the groups. Data are presented as mean 
± S.D (N = 6), ***P < 0.001 versus vehicle-treated group. Precaution was taken to avoid necrotic area during the Ki measurement by com-
paring the QAR brain section with a corresponding H&E stained serial brain tumor section.



205

Targeted Brain Tumor Treatment

Drug Target Insights 2007: 2 

TMZ, selectively to reach brain tumor and brain 
tissue surrounding tumor, which represents prolif-
erating edges of tumor where the BBB may be 
intact (Pardridge et al. 1992). Furthermore, we 
showed that Trastuzumab combined with TMZ 
co-administered with potassium channel agonists 
signifi cantly increased survival rates in mice with 
intracranial GBM xenograft (unpublished data). 
These results are consistent with our earlier study, 
where we showed that potassium channel activator 
(Minoxidil sulfate: MS) infusion selectively 
enhanced carboplatin delivery to tumor tissue 
without increasing delivery to normal brain 
(Ningaraj, Rao and Black, 2003b). MS co-infusion 
with carboplatin in rats resulted in tumor regression, 
significantly increasing survival (Black and 
Ningaraj, 2004). The ability to deliver Her-2 neu 
targeting drug Herceptin (Trastuzumab) by potas-
sium channel-mediated BTB modulation in human 
xenografts may be clinically useful because GBMs 
frequently have altered receptor tyrosine kinase 
genes (Fuller and Bigner, 1992), including Her-2 
neu that is over expressed in about 17%–20% of 
GBM patients (Forseen et al. 2002) resulting in 
poor prognosis and patient survival. A molecular 
target-based therapy using Trastuzumab and 
Pertuzumab (Omnitarg, 2C4) is developed by 
Genentech Inc., for brain tumor, but their delivery 
across the BTB remains a major concern.

Molecular medicine 
To conclude, the future of molecular targeted 
therapy is to achieve customized treatment strategy 
for brain tumor patients, where individual patient 
treatment will be based on the molecular profi le of 
the disease. The information based on the changing 
levels of active genes/proteins inside tumor cells in 
response to an anti-cancer drug, could help physi-
cians to determine early in the treatment whether a 
drug works effectively or not. Researchers have 
identifi ed gene/protein markers that are useful in 
individualizing treatment in prostate, breast, and 
ovarian cancer patients. Although, brain tumor tissue 
is heterogeneous, the genetic profi ling of tumor 
tissue gives valuable molecular information. As a 
case in point, high-throughput gene profi ling of 
brain tumor biopsy samples by gene array technique 
can be compared with genomic data generated using 
tumor samples to predict whether patients would 
benefit from anti-cancer drug (like Gleevec) 
treatment or by potassium channel modulation in 
tumor and tumor vascular endothelial cells.

Acknowledgments
We thank Robert Bishop, Ph.D., Schering-Plough 
Research Institute, Kenilworth, New Jersey for 
kindly providing radiolabeled and non-radiolabeled 
Temozolomide. We also acknowledge American 
Cancer Society award to NSN.

References
Ambros, V. 2003. MicroRNA pathways in fl ies and worms: growth, death, 

fat, stress, and timing. Cell, 113:673–6.
Alaminos, M. et al. 2005. EMP3, a myelin-related gene located in the 

critical 19q13.3 region, is epigenetically silenced and exhibits features 
of a candidate tumor suppressor in glioma and neuroblastoma. 
Cancer Res., 65:2565–71.

Bachman, K.E. et al. 1999. Methylation-associated silencing of the tissue 
inhibitor of metalloproteinase-3 gene suggest a suppressor role in 
kidney, brain, and other human cancers. Cancer Res., 59:798–802.

Balana, C.et al. 2004. Phase II study of temozolomide and cisplatin as primary 
treatment prior to radiotherapy in newly diagnosed glioblastoma multi-
forme patients with measurable disease. A study of the Spanish Medical 
Neuro-Oncology Group (GENOM). J. Neurooncol., 70:359–69.

Bartel, B. and Bartel, D.P. 2003. MicroRNAs: at the root of plant develop-
ment? Plant Physiol., 132:709–17.

Bartel, D.P. 2004. MicroRNAs: genomics, biogenesis, mechanism, and 
function. Cell, 116:281–97.

Baumann, F. et al. 2004. Combined thalidomide and temozolomide treat-
ment in patients with glioblastoma multiforme. J. Neurooncol., 
67:191–200.

Baylin, S.B. et al. 2001. Aberrant patterns of DNA methylation, chromatin forma-
tion and gene expression in cancer. Hum. Mol. Genet., 10:687–92.

Bernsen, H.J. and van der Kogel, A.J. 1999. Antiangiogenic therapy in brain 
tumor models. J. Neurooncol., 45:247–55.

Bergers, G. et al. 2003. Benefi ts of targeting both pericytes and endothelial 
cells in the tumor vasculature with kinase inhibitors. J. Clin. Invest., 
111:1287–95.

Belinsky, S.A. et al. 2006. Promoter hypermethylation of multiple genes in 
sputum precedes lung cancer incidence in a high-risk cohort. Cancer 
Res., 66:3338–44.

Black, K.L. and Ningaraj, N.S. 2004. Modulation of brain tumor capillaries 
for enhanced drug delivery selectively to brain tumor. Cancer Control, 
11:165–73.

Bocangel, D.B. et al. 2002. Multifaceted resistance of gliomas to temozolo-
mide. Clin. Cancer Res., 8:2725–34.

Butowski, N. and Chang, S.M. 2005. Small molecule and monoclonal an-
tibody therapies in neurooncology. Cancer Control, 12:116–24.

Cameron, E.E. et al. 1999. Synergy of demethylation and histone deacety-
lase inhibition in the re-expression of genes silenced in cancer. Nat. 
Genet., 21:103–7.

Cairns, P. et al. 2001. Molecular detection of prostate cancer in urine by 
GSTP1 hypermethylation. Clin. Cancer Res., 7:2727–30.

Calin, G.A. and Croce, C.M. 2006a. Genomics of chronic lymphocytic 
leukemia microRNAs as new players with clinical signifi cance. 
Semin. Oncol., 33:167–73.

Calin, G.A. and Croce, C.M. 2006b. MicroRNAs and chromosomal abnor-
malities in cancer cells. Oncogene, 25:6202–10.

Chan, J.A., Krichevsky, A.M. and Kosik, K.S. 2005. MicroRNA-21 is an 
antiapoptotic factor in human glioblastoma cells. Cancer Res., 
65:6029–33.

Ciafre, S.A. et al. 2005. Extensive modulation of a set of microRNAs in primary 
glioblastoma. Biochem. Biophys. Res. Commun., 334:1351–8.

Costello, J.F. et al. 1996. Silencing of p16/CDKN2 expression in human 
gliomas by methylation and chromatin condensation. Cancer Res., 
56:2405–10.

Costello, J.F. et al. 2000. Aberrant CpG-island methylation has non-random 
and tumour-type-specifi c patterns. Nat. Genet., 24:132–8.



206

Ningaraj et al

Drug Target Insights 2007: 2

Costello, J.F. 2003. DNA methylation in brain development and glioma-
genesis. Front Biosci., 8:s175–84.

Dalmay, T. and Edwards, D.R. 2006. MicroRNAs and the hallmarks of 
cancer. Oncogene, 25:6170–5.

Daskalakis, M. et al. 2002. Demethylation of a hypermethylated P15/INK4B 
gene in patients with myelodysplastic syndrome by 5-Aza-2′-
deoxycytidine (decitabine) treatment. Blood, 100:2957–64.

Fabian, M.A. et al. 2005. A small molecule-kinase interaction map for 
clinical kinase inhibitors. Nat. Biotechnol., 23:329–36.

Forseen, S.E. et al. 2002. Identifi cation and relationship of HER-2/neu 
overexpression to short-term mortality in primary malignant brain 
tumors. Anticancer Res., 22:1599–602.

Foltz, G. et al. 2006. Genome-wide analysis of epigenetic silencing identi-
fi es BEX1 and BEX2 as candidate tumor suppressor genes in malig-
nant glioma. Cancer Res., 66:6665–74.

Friedman, H.S., Kerby, T. and Calvert, H. 2000. Temozolomide and treat-
ment of malignant glioma. Clin. Cancer Res., 6:2585–97.

Fuller, G.N. and Bigner, S.H. 1992. Amplifi ed cellular oncogenes in neoplasms 
of the human central nervous system. Mutat. Res., 276:299–306.

Fueyo, J. et al. 1996. Hypermethylation of the CpG island of p16/CDKN2 
correlates with gene inactivation in gliomas. Oncogene, 
13:1615–9.

Gullick, W.J. 2001. Update on HER-2 as a target for cancer therapy: alter-
native strategies for targeting the epidermal growth factor system in 
cancer. Breast Cancer Res., 3:390–4.

Goffi n, J. and Eisenhauer, E. 2002. DNA methyltransferase inhibitors-state 
of the art. Ann. Oncol., 13:1699–716.

Gurney, A.M. and Hunter, E. 2005. The use of small interfering RNA to 
elucidate the activity and function of ion channel genes in an intact 
tissue. J. Pharmacol. Toxicol. Methods, 51:253–62.

Hegi, M.E. et al. 1997. Hemizygous or homozygous deletion of the chromo-
somal region containing the p16INK4a gene is associated with amplifi cation 
of the EGF receptor gene in glioblastomas. Int. J. Cancer., 73:57–63.

Hirose, Y. Berger, M.S. and Pieper, R.O. 2001. p53 effects both the duration 
of G2/M arrest and the fate of temozolomide-treated human glioblas-
toma cells. Cancer Res., 61:1957–63.

Hutvagner, G. 2006. MicroRNAs and cancer: issue summary. Oncogene, 
25:6154–5.

Ichimura, K. et al. 1996. Human glioblastomas with no alterations of the 
CDKN2A (p16INK4A, MTS1) and CDK4 genes have frequent 
mutations of the retinoblastoma gene. Oncogene., 13:1065–72.

Ichimura, K. et al. 2000. Deregulation of the p14ARF/MDM2/p53 pathway 
is a prerequisite for human astrocytic gliomas with G1-S transition 
control gene abnormalities. Cancer Res., 60:417–24.

Jannot, G. and Simard, M.J. 2006. Tumour-related microRNAs functions 
in Caenorhabditis elegans. Oncogene, 25:6197–201.

Jovanovic, M. and Hengartner, M.O. 2006. miRNAs and apoptosis: RNAs 
to die for. Oncogene, 25:6176–87.

Kanzawa, T. et al. 2003. Inhibition of DNA repair for sensitizing resistant 
glioma cells to temozolomide. J. Neurosurg., 99:1047–52.

Kent, O.A. and Mendell, J.T. 2006. A small piece in the cancer puzzle: microRNAs 
as tumor suppressors and oncogenes. Oncogene, 25:6188–96.

Kim, T.Y. et al. 2006. Epigenomic profi ling reveals novel and frequent 
targets of aberrant DNA methylation-mediated silencing in malignant 
glioma. Cancer Res., 66:7490–501.

Komine, C. et al. 2003. Promoter hypermethylation of the DNA repair gene 
O6-methylguanine-DNA methyltransferase is an independent predic-
tor of shortened progression free survival in patients with low grade 
diffuse astrocytomas. Brain Pathol., 13:176–84.

Kraft, R. et al. 2003. BK channel openers inhibit migration of human glio-
ma cells. Pfl ugers Arch., 446:248–55.

Leis, J.F. et al. 2004. Central nervous system failure in patients with 
chronic myelogenous leukemia lymphoid blast crisis and Philadelphia 
chromosome positive acute lymphoblastic leukemia treated with 
imatinib (STI-571). Leuk Lymphoma., 45:695–8.

Levin, V.A. 2007. Are gliomas preventable? Recent Results Cancer Res., 
174:205–5.

Libermann, T.A. et al. 1985. Amplifi cation, enhanced expression and pos-
sible rearrangement of EGF receptor gene in primary human brain 
tumours of glial origin. Nature, 313:144–7.

Lund, E.L., Bastholm, L. and Kristjansen, P.E. 2000. Therapeutic synergy 
of TNP-470 and ionizing radiation: effects on tumor growth, vessel 
morphology, and angiogenesis in human glioblastoma multiforme 
xenografts. Clin. Cancer Res., 6:971–8.

Lubbert, M. 2000. DNA methylation inhibitors in the treatment of leuke-
mias, myelodysplastic syndromes and hemoglobinopathies: clinical 
results and possible mechanisms of action. Curr. Top Microbiol. 
Immunol., 249:135–64.

Mashiyama, S. et al. 1991. Detection of p53 gene mutations in human brain 
tumors by single-strand conformation polymorphism analysis of 
polymerase chain reaction products. Oncogene., 6:1313–8.

Mauceri, H.J. et al. 1998. Combined effects of angiostatin and ionizing 
radiation in antitumour therapy. Nature, 394:287–91.

Ma, H.I. et al. 2002. Intratumoral gene therapy of malignant brain tumor in 
a rat model with angiostatin delivered by adeno-associated viral 
(AAV) vector. Gene Ther., 9:2–11.

Mellinghoff, I.K. et al. 2005. Molecular determinants of the response of 
glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med., 
353:2012–24.

Nakamura, M. et al. 1996. Promoter hypermethylation of the RB1 gene in 
glioblastomas. Lab Invest., 81:77–82.

Newton, H.B. 2003. Molecular neuro-oncology and development of 
targeted therapeutic strategies for brain tumors. Part 1: Growth 
factor and Ras signaling pathways. Expert Rev. Anticancer Ther., 
3:595–614.

Ningaraj, N.S. et al. 2002. Regulation of blood-brain tumor barrier 
permeability by calcium-activated potassium channels. J. Pharmacol. 
Exp. Ther., 301:838–51.

Ningaraj, N.S., Rao, M.K. and Black, K.L. 2003a. Adenosine 5′-triphos-
phate-sensitive potassium channel-mediated blood-brain tumor 
barrier permeability increase in a rat brain tumor model. Cancer 
Res., 63:8899–911.

Ningaraj, N.S. 2006. Drug delivery to brain tumours: challenges and prog-
ress. Expert Opin. Drug Deliv., 3:499–509.

O’Driscoll L. 2006. The emerging world of microRNAs. Anticancer Res., 
26:4271–8.

Olsen, M.L., Weaver, A.K., Ritch, P.S., Sontheimer, H. 2004. Modulation 
of glioma BK channels via erbB2, 81:2179–89.

Osada, H. and Takahashi, T. 2006. MicroRNAs in biological processes and 
carcinogenesis. Carcinogenesis, 28:2–12.

Pardridge, W.M. et al. 1992. Blood-brain barrier and new approaches to 
brain drug delivery. West J. Med., 156:281–6.

Pardridge, W.M. 2001. Drug targeting, drug discovery, and brain drug 
development. In: Brain drug targeting: The future of brain drug devel-
opment. Cambridge: Cambridge University Press, pp. 1–13.

Plumb, J.A. et al. 2000. Reversal of drug resistance in human tumor xeno-
grafts by 2′-deoxy-5-azacytidine-induced demethylation of the 
hMLH1 gene promoter. Cancer Res., 60:6039–44.

Pollack, I.F. et al. 2007. Phase I trial of imatinib in children with newly 
diagnosed brainstem and recurrent malignant gliomas: a Pediatric 
Brain Tumor Consortium report. Neuro-oncol., 9:145–60.

Rao, M. and Ningaraj, N. 2001. Activation of Calcium-dependent potas-
sium channels elicits selective glioma cell death. In: AACR-NCI-
EORTC.

Rege, T.A., Fears, C.Y. and Gladson, C.L. 2005. Endogenous inhibitors of 
angiogenesis in malignant gliomas: nature’s antiangiogenic therapy. 
Neuro-oncol., 7:106–21.

Reijneveld, J.C., Voest, E.E. and Taphoorn, M.J. 2000. Angiogenesis in malig-
nant primary and metastatic brain tumors. J. Neurol., 247:597–608.

Reardon, D.A. et al. 2005. Phase II study of imatinib mesylate plus 
hydroxyurea in adults with recurrent glioblastoma multiforme. 
J. Clin. Oncol., 23:9359–68.

Rich, J.N. and Bigner, D.D. 2004. Development of novel targeted therapies in 
the treatment of malignant glioma. Nat. Rev. Drug Discov., 3:430–46.



207

Targeted Brain Tumor Treatment

Drug Target Insights 2007: 2 

Robinson, S.P. et al. 2003. Tumour dose response to the antivascular agent 
ZD6126 assessed by magnetic resonance imaging. Br. J. Cancer., 
88:1592–7.

Saito, Y. and Jones, P.A. 2006. Epigenetic Activation of Tumor Suppressor 
MicroRNAs in Human Cancer Cells. Cell Cycle, 5:2220–22.

Shusterman, S. et al. 2001. The angiogenesis inhibitor tnp-470 effectively 
inhibits human neuroblastoma xenograft growth, especially in the 
setting of subclinical disease. Clin. Cancer Res., 7:977–84.

Stupp, R. et al. 2005. Radiotherapy plus concomitant and adjuvant temo-
zolomide for glioblastoma. N. Engl. J. Med., 352:987–96.

Takano, S. et al. 2004. Angiogenesis and antiangiogenic therapy for malig-
nant gliomas. Brain Tumor Pathol., 21:69–73.

Thomas, M. 2003. Epidermal growth factor receptor tyrosine kinase inhibitors: 
application in non-small cell lung cancer. Cancer Nurs., 26: 21S–25S.

Weaver, A.K., Bomben, V.C. and Sontheimer, H. 2006. Expression and 
function of calcium-activated potassium channels in human glioma 
cells. Glia., 54:223–33.

Weaver, A.K., Liu, X. and Sontheimer, H. 2004. Role for calcium-activated 
potassium channels (BK) in growth control of human malignant 
glioma cells. J. Neurosci. Res., 78:224–34.

Wen, P.Y. et al. 2006. Phase I/II study of imatinib mesylate for recurrent 
malignant gliomas: North American Brain Tumor Consortium Study 
99–08. Clin. Cancer Res., 12:4899–907.

Yamanaka, R. et al. 2003. Induction of an antitumor immunological response 
by an intratumoral injection of dendritic cells pulsed with geneti-
cally engineered Semliki Forest virus to produce interleukin-18 
combined with the systemic administration of interleukin-12. 
J. Neurosurg., 99:746–53.

Yang, W. et al. 2005. Development of a syngeneic rat brain tumor model 
expressing EGFRvIII and its use for molecular targeting studies with 
monoclonal antibody L8A4. Clin. Cancer Res., 11:341–50.

Yung, W.K. et al. 1999. Multicenter phase II trial of temozolomide in 
patients with anaplastic astrocytoma or anaplastic oligoastrocy-
toma at first relapse. Temodal Brain Tumor Group. J. Clin. Oncol., 
17:2762–71.

Zhang, L. and Coukos, G. 2006. MicroRNAs: A New Insight into Cancer 
Genome. Cell Cycle, 5:2216–19.

















<<
  /ASCII85EncodePages false
  /AllowTransparency false
  /AutoPositionEPSFiles true
  /AutoRotatePages /None
  /Binding /Left
  /CalGrayProfile (Dot Gain 20%)
  /CalRGBProfile (sRGB IEC61966-2.1)
  /CalCMYKProfile (U.S. Web Coated \050SWOP\051 v2)
  /sRGBProfile (sRGB IEC61966-2.1)
  /CannotEmbedFontPolicy /Error
  /CompatibilityLevel 1.4
  /CompressObjects /Tags
  /CompressPages true
  /ConvertImagesToIndexed true
  /PassThroughJPEGImages true
  /CreateJDFFile false
  /CreateJobTicket false
  /DefaultRenderingIntent /Default
  /DetectBlends true
  /ColorConversionStrategy /LeaveColorUnchanged
  /DoThumbnails false
  /EmbedAllFonts true
  /EmbedJobOptions true
  /DSCReportingLevel 0
  /EmitDSCWarnings false
  /EndPage -1
  /ImageMemory 1048576
  /LockDistillerParams false
  /MaxSubsetPct 100
  /Optimize true
  /OPM 1
  /ParseDSCComments true
  /ParseDSCCommentsForDocInfo true
  /PreserveCopyPage true
  /PreserveEPSInfo true
  /PreserveHalftoneInfo false
  /PreserveOPIComments false
  /PreserveOverprintSettings true
  /StartPage 1
  /SubsetFonts true
  /TransferFunctionInfo /Apply
  /UCRandBGInfo /Preserve
  /UsePrologue false
  /ColorSettingsFile ()
  /AlwaysEmbed [ true
  ]
  /NeverEmbed [ true
  ]
  /AntiAliasColorImages false
  /DownsampleColorImages true
  /ColorImageDownsampleType /Bicubic
  /ColorImageResolution 300
  /ColorImageDepth -1
  /ColorImageDownsampleThreshold 1.50000
  /EncodeColorImages true
  /ColorImageFilter /DCTEncode
  /AutoFilterColorImages true
  /ColorImageAutoFilterStrategy /JPEG
  /ColorACSImageDict <<
    /QFactor 0.15
    /HSamples [1 1 1 1] /VSamples [1 1 1 1]
  >>
  /ColorImageDict <<
    /QFactor 0.15
    /HSamples [1 1 1 1] /VSamples [1 1 1 1]
  >>
  /JPEG2000ColorACSImageDict <<
    /TileWidth 256
    /TileHeight 256
    /Quality 30
  >>
  /JPEG2000ColorImageDict <<
    /TileWidth 256
    /TileHeight 256
    /Quality 30
  >>
  /AntiAliasGrayImages false
  /DownsampleGrayImages true
  /GrayImageDownsampleType /Bicubic
  /GrayImageResolution 300
  /GrayImageDepth -1
  /GrayImageDownsampleThreshold 1.50000
  /EncodeGrayImages true
  /GrayImageFilter /DCTEncode
  /AutoFilterGrayImages true
  /GrayImageAutoFilterStrategy /JPEG
  /GrayACSImageDict <<
    /QFactor 0.15
    /HSamples [1 1 1 1] /VSamples [1 1 1 1]
  >>
  /GrayImageDict <<
    /QFactor 0.15
    /HSamples [1 1 1 1] /VSamples [1 1 1 1]
  >>
  /JPEG2000GrayACSImageDict <<
    /TileWidth 256
    /TileHeight 256
    /Quality 30
  >>
  /JPEG2000GrayImageDict <<
    /TileWidth 256
    /TileHeight 256
    /Quality 30
  >>
  /AntiAliasMonoImages false
  /DownsampleMonoImages true
  /MonoImageDownsampleType /Bicubic
  /MonoImageResolution 1200
  /MonoImageDepth -1
  /MonoImageDownsampleThreshold 1.50000
  /EncodeMonoImages true
  /MonoImageFilter /CCITTFaxEncode
  /MonoImageDict <<
    /K -1
  >>
  /AllowPSXObjects false
  /PDFX1aCheck false
  /PDFX3Check false
  /PDFXCompliantPDFOnly false
  /PDFXNoTrimBoxError true
  /PDFXTrimBoxToMediaBoxOffset [
    0.00000
    0.00000
    0.00000
    0.00000
  ]
  /PDFXSetBleedBoxToMediaBox true
  /PDFXBleedBoxToTrimBoxOffset [
    0.00000
    0.00000
    0.00000
    0.00000
  ]
  /PDFXOutputIntentProfile ()
  /PDFXOutputCondition ()
  /PDFXRegistryName (http://www.color.org)
  /PDFXTrapped /Unknown

  /Description <<
    /JPN <FEFF3053306e8a2d5b9a306f30019ad889e350cf5ea6753b50cf3092542b308030d730ea30d730ec30b9537052377528306e00200050004400460020658766f830924f5c62103059308b3068304d306b4f7f75283057307e305930023053306e8a2d5b9a30674f5c62103057305f00200050004400460020658766f8306f0020004100630072006f0062006100740020304a30883073002000520065006100640065007200200035002e003000204ee5964d30678868793a3067304d307e305930023053306e8a2d5b9a306b306f30d530a930f330c8306e57cb30818fbc307f304c5fc59808306730593002>
    /FRA <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>
    /DEU <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>
    /PTB <FEFF005500740069006c0069007a006500200065007300740061007300200063006f006e00660069006700750072006100e700f5006500730020007000610072006100200063007200690061007200200064006f00630075006d0065006e0074006f0073002000500044004600200063006f006d00200075006d00610020007200650073006f006c007500e700e3006f00200064006500200069006d006100670065006d0020007300750070006500720069006f0072002000700061007200610020006f006200740065007200200075006d00610020007100750061006c0069006400610064006500200064006500200069006d0070007200650073007300e3006f0020006d0065006c0068006f0072002e0020004f007300200064006f00630075006d0065006e0074006f0073002000500044004600200070006f00640065006d0020007300650072002000610062006500720074006f007300200063006f006d0020006f0020004100630072006f006200610074002c002000520065006100640065007200200035002e00300020006500200070006f00730074006500720069006f0072002e00200045007300740061007300200063006f006e00660069006700750072006100e700f50065007300200072006500710075006500720065006d00200069006e0063006f00720070006f0072006100e700e3006f00200064006500200066006f006e00740065002e>
    /DAN <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>
    /NLD <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>
    /ESP <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>
    /SUO <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>
    /ITA <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>
    /NOR <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>
    /SVE <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>
    /ENU <FEFF005500730065002000740068006500730065002000730065007400740069006e0067007300200074006f0020006300720065006100740065002000500044004600200064006f00630075006d0065006e0074007300200077006900740068002000680069006700680065007200200069006d0061006700650020007200650073006f006c007500740069006f006e00200066006f0072002000680069006700680020007100750061006c0069007400790020007000720065002d007000720065007300730020007000720069006e00740069006e0067002e0020005400680065002000500044004600200064006f00630075006d0065006e00740073002000630061006e0020006200650020006f00700065006e00650064002000770069007400680020004100630072006f00620061007400200061006e0064002000520065006100640065007200200035002e003000200061006e00640020006c0061007400650072002e002000540068006500730065002000730065007400740069006e006700730020007200650071007500690072006500200066006f006e007400200065006d00620065006400640069006e0067002e>
  >>
>> setdistillerparams
<<
  /HWResolution [2400 2400]
  /PageSize [612.000 792.000]
>> setpagedevice