ArticlePDF Available

Novel Therapies in Glioblastoma

Wiley
Neurology Research International
Authors:

Abstract and Figures

Conventional treatment of glioblastoma has advanced only incrementally in the last 30 years and still yields poor outcomes. The current strategy of surgery, radiation, and chemotherapy has increased median survival to approximately 15 months. With the advent of molecular biology and consequent improved understanding of basic tumor biology, targeted therapies have become cornerstones for cancer treatment. Many pathways (RTKs, PI3K/AKT/mTOR, angiogenesis, etc.) have been identified in GBM as playing major roles in tumorigenesis, treatment resistance, or natural history of disease. Despite the growing understanding of the complex networks regulating GBM tumors, many targeted therapies have fallen short of expectations. In this paper, we will discuss novel therapies and the successes and failures that have occurred. One clear message is that monotherapies yield minor results, likely due to functionally redundant pathways. A better understanding of underlying tumor biology may yield insights into optimal targeting strategies which could improve the overall therapeutic ratio of conventional treatments.
Content may be subject to copyright.
Hindawi Publishing Corporation
Neurology Research International
Volume 2012, Article ID 428565, 14 pages
doi:10.1155/2012/428565
Review Article
Novel Therapies in Glioblastoma
James Perry, Masahiko Okamoto, Michael Guiou, Katsuyuki Shirai, Allison Errett,
and Arnab Chakravarti
Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute,
The Ohio State University, Columbus, OH 43210, USA
Correspondence should be addressed to James Perry, james.perry@osumc.edu
Received 16 August 2011; Accepted 9 December 2011
Academic Editor: Stuart Burri
Copyright © 2012 James Perry et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Conventional treatment of glioblastoma has advanced only incrementally in the last 30 years and still yields poor outcomes. The
current strategy of surgery, radiation, and chemotherapy has increased median survival to approximately 15 months. With the
advent of molecular biology and consequent improved understanding of basic tumor biology, targeted therapies have become
cornerstones for cancer treatment. Many pathways (RTKs, PI3K/AKT/mTOR, angiogenesis, etc.) have been identified in GBM as
playing major roles in tumorigenesis, treatment resistance, or natural history of disease. Despite the growing understanding of
the complex networks regulating GBM tumors, many targeted therapies have fallen short of expectations. In this paper, we will
discuss novel therapies and the successes and failures that have occurred. One clear message is that monotherapies yield minor
results, likely due to functionally redundant pathways. A better understanding of underlying tumor biology may yield insights into
optimal targeting strategies which could improve the overall therapeutic ratio of conventional treatments.
1. Introduction
Glioblastoma (GBM) is a grade IV glioma and accounts
for approximately 75% of all high-grade gliomas with
approximately 9,000 new cases per year diagnosed in the
United States alone, making it the most common adult
brain tumor. GBM is the most aggressive glial neoplasm,
and despite advances in medical management, the out-
comes remain quite poor. The current standard of care for
high-grade glioma patients is maximum surgical resection
combined with radiation and concomitant and adjuvant
temozolomide (TMZ) therapy. The addition of radiotherapy
for the treatment of GBMs led to the first significant
improvement in patient survival starting in the late 1970s.
More recently, Stupp et al. have shown that the addition
of the chemotherapeutic agent TMZ can increase survival
further to approximately 15 months.
2. Development of Standard of Care (RT + TMZ)
Surgery is a critical component of standard of care, allowing
histological diagnosis, but more critically, tumor debulking.
This greatly reduces the number of cells to be killed by
radiation or chemotherapy. It also decreases intracranial
pressure which, depending on the location of the tumor, may
result in recovery of CNS function or decrease in usage of
corticosteroid. Recently, the eectiveness of aggressive surgi-
cal resection on survival was suggested by some prospective
analyses [13].
Unfortunately, most glioblastomas recur following
surgery. The ecacy of radiation therapy (RT) was reported
in the 1970s [4,5], and RT has become a standard adjuvant
therapy in patients with malignant glioma. In 2005, the
eciency of concomitant and adjuvant TMZ was suggested
by a phase III study that was conducted by the European
Organization for Research and Treatment of Cancer
(EORTC) and the National Cancer Institute of Canada
(NCIC) [6]. In the EORTC/NCIC study, a total of 573
patients with newly diagnosed glioblastoma were enrolled.
The authors reported the combined therapy of TMZ and RT
increased median survival time (MST) when compared with
RT-alone (14.6 months versus 12.1 months, P<.001). At the
5-year analysis of this study, the 5-year overall survival rate
was 9.8% for the combination therapy group versus 1.9%
2Neurology Research International
for the RT alone group (P<.001), with a median follow-up
of 61 months [7]. With this strong evidence, combination
therapy with TMZ and RT is widely prescribed and currently
considered the standard treatment for patients with newly
diagnosed glioblastoma.
Some investigators had suggested that the epigenetic
silencing of a DNA repair enzyme named O-6-methylgua-
nine-DNA methyltransferase (MGMT) by promoter methy-
lation was associated with good prognosis for patients with
glioblastoma treated with alkylating agents such as TMZ
[8,9]. In agreement with previous studies, patients with
a methylated MGMT promoter had significantly improved
MST when compared with patients with an unmethylated
MGMT promoter (21.7 months versus 15.3 months, P<
.001) in the EORTC/NCIC trial [10]. Furthermore, this
study indicated that patients with an unmethylated MGMT
promoter received less benefit from the combined therapy.
MGMT promoter methylation status is widely used to
predict the ecacy for combination therapy of RT and TMZ
for newly diagnosed glioblastoma.
Although the combination therapy of RT and TMZ
has become standard, most patients will still eventually
recur. Thus the development of a new treatment strategy is
needed in order to overcome the resistance of glioblastoma
to current therapy. One strategy is increasing the intensity
of radiation dose. However, neither radiosurgery boost
[11] nor brachytherapy boost [12] shows improvement
in survival. Another strategy is the optimization of TMZ
usage by approaches such as dose-dense regimens. RTOG
0525/EORTC 26052-22053, a prospective randomized trial,
was conducted by the Radiation Therapy Oncology Group
International (RTOG) and EORTC. It aimed to determine
whether a dose-dense TMZ regimen is more eective than
the standard TMZ regimen in the adjuvant setting, and the
results of this study showed no significant dierence between
the two arms [13]. Moreover, the adverse eects, especially
in the field of lymphopenia and fatigue, were significantly
increased in the dose-dense arm.
3. Technological Advancements in
GBM Therapy
Current research eorts in both the basic and clinical sciences
are improving clinicians’ ability to more accurately target and
treat GBM. In addition to targeted therapies, which will be
discussed later in this paper, there have been advancements
in the technological arena that are improving patient care.
3.1. Target Delineation. MRI remains the gold standard
for delineating tumor in both the pre- and postoperative
setting. Gross tumor volume is felt to be best represented
by areas of contrast-enhanced T1 signal while areas of
T2/FLAIR enhancement reflect regions of infiltrative tumor.
These volumes form the basis for radiotherapy target
delineation. A limitation of contrast-enhanced MRI is that
it relies on surrogate markers of tumor presence (tumor-
associated breakdown of the blood-brain barrier and cerebral
edema) versus a direct measure of actual gross tumor
mass and spread. Functional imaging techniques such as
positron emission tomography (PET) play a pivotal role
in the staging and planning of cancers in other parts
of the body. Unfortunately, the most widely used radio-
tracer [18F]-fluorodeoxyglucose is relatively insensitive at
delineating malignancy in the brain due to the high basal
metabolic rate of normal brain tissue [14]. Other clini-
cally available radiotracers such as L-methyl-11C-methionine
(MET) and O-(2-[18F]fluoroethyl)-L-tyrosine(FET) have
shown promise in localizing gliomas. Recent studies have
shown MET PET can more accurately identify areas of active
tumor versus traditional MRI [15,16]. MET is actively taken
up by gliomas but shows only low uptake in normal brain. In
a study of 14 patients with high-grade glioma, MET PET was
highly correlated with areas of endothelial proliferation and
mitotic activity [17], a more direct marker of tumor activity
that can be visualized using MRI. In a study of 26 patients
with GBM, Lee et al. [15] showed that 5 of 26 patients had
areas of MET PET positivity outside radiotherapy volumes
defined by MRI. All of these patients had noncentral failures.
In 14 patients where MET PET-positive areas were covered
in the high-dose radiotherapy volume, none had noncentral
failures. FET PET has also been shown to correlate with
areas of active tumor. In a study of 31 patients with GBM,
FET PET was highly correlated with areas of active tumor
on biopsy [18]. FET shows minimal uptake in macrophages
or inflammatory tissues, indicating that it may be superior
to MET in identifying areas of active tumor, especially in
the postoperative setting. In a study comparing FET PET to
MRI, Piroth et al. showed that areas active on FET PET not
covered by MRI-based radiotherapy target volumes predict
a shorter disease-free survival (5.1 versus 9.6 months) and
overall survival (6.9 versus 20 months) further supporting
the integration of metabolically based treatment planning
methods into radiation planning for GBM [19]. An emerging
technique that may also play an important role in target
delineation for GBM is diusion tensor imaging (DTI). DTI
uses specialized MRI sequences to measure the diusion of
water in the brain, the greatest values of which lie along white
matter tracts. Postmortem studies in GBM patients show
that glioma cells tend to migrate the greatest distance from
the primary site along these tracts. In a study of 14 patients
with recurrent GBM, Krishnan et al. were able to show a
strong relationship between the sites of recurrence and the
DTI maps emanating from the primary site [20]. While none
of the aforementioned imaging techniques have been tested
in a phase III study, they provide promising tools by which
clinicians cannot only more accurately identify areas of active
tumor but also potentially predict their most likely route of
spread to help refine radiotherapy treatment fields.
3.2. Disease Monitoring. Treatment-related eects such as
postoperative scarring and hemorrhage, peritumoral edema,
inflammation, and microvascular changes make radio-
graphic assessment problematic as these changes can mimic
progressive disease. This has been termed pseudoprogres-
sion. Additionally, given the high doses and often large vol-
umes required to treat GBM, up to 30% of patients develop
radionecrosis which can also mimic disease recurrence on
Neurology Research International 3
MRI [21]. The front-line treatment for radionecrosis is
steroids and time. Some patients require surgical resection
for more advanced cases resistant to steroid treatment. The
ability to diagnose true progression and initiate second-line
therapy is paramount in a patient population with such
poor disease-free and overall survival. Both PET imaging and
magnetic resonance spectroscopy (MRS) have been shown
to be superior to MRI in helping distinguish active disease
from pseudoprogression or radionecrosis. The specificity
of FET PET and MET PET to detect recurrent disease is
approximately 90% and 70%, respectively [19]. Areas active
on FET PET show a high correlation with biopsy-proven
areas of active disease [18], and regions of enhancing tissue
on MRI are often negative on FET PET. Numerous studies
of magnetic resonance spectroscopy (MRS), which measures
the dierential concentrations of metabolites in the brain,
also show improved accuracy in distinguishing recurrent
disease versus MRI (71% versus 55%) [22]. Classic MRS
is hampered by poor spatial resolution and nonvolumetric
data; however, recent advances in technology now permit
volumetric MRS which may improve its diagnostic value.
Integration of these metabolically based imaging techniques
may help to improve our ability to detect disease progression
early and optimize 2nd-line therapy for patients.
3.3. Treatment in the Recurrent Setting. The vast majority
of patients with GBM recur within 8–12 months follow-
ing completion of therapy highlighting the importance of
developing ecacious treatments for patients with recurrent
disease. Studies examining reirradiation with or without
chemotherapy have shown the most promising results. Given
the fact that the majority of patients fail within 2 cm of
the primary site, numerous studies examining the ecacy
of stereotactic radiosurgery (SRS), as delivered by a variety
of dierent methods, have been completed [2325]. SRS
provides a modest improvement in outcome but with
significant risk of radionecrosis requiring surgical resection.
Patients undergoing fractionated stereotactic radiotherapy
(FSRT) at the time of recurrence have median survival
ranging from approximately 7 to 14 months for all patients
but with reduced rates of morbidity [23,2628]. Dosing
regimens with a cumulative dose exceeding 40Gy appear to
be associated with a greater degree of radiation damage [29].
Response on postprocedure MRI seems to be an important
prognostic factor for either SRS or FSRT. In a study of 36
patients (26 SRS, 10 FSRT) patients showing response on
posttreatment MRI had median survival of 15.8 months
versus 7.3 months for nonresponders [23].
Combination chemoradiotherapy has shown some e-
cacy in the recurrent setting. In a study of 25 patients with
recurrent high-grade glioma (20 GBM, 4 anaplastic astro-
cytoma), treatment with FSRT (30 Gy in 5 fractions) plus
concurrent bevacizumab resulted in posttreatment median
survival of 12.5 months [30] with comparable toxicity
rates as reported by other studies of GBM patients treated
with bevacizumab. Three of 25 patients had to discontinue
treatment due to CNS toxicity, wound complications, and
bowel perforation. A recent phase I dose-escalation trial
of gefitinib, an epidermal growth factor receptor (EGFR)
tyrosine kinase inhibitor (TKI), plus FSRT (18 to 36 Gy in 3
fractions) in 15 patients (11 GBM, 4 anaplastic astrocytoma)
showed this combination was well tolerated at all dose levels.
Median progression-free survival was 7 months with a 6-
month progression-free survival of 63% and a 1-year overall
survival of 40%. Of course, given the mixture of grade III
and grade IV histologies, it is dicult to compare outcomes
with studies of GBM patients alone, but these initial results
show promise. Studies combining cytotoxic chemotherapy
with FSRT agents have shown similar response rates with
acceptable toxicity profiles [3133].
Recurrent malignant gliomas can also be managed with
chemotherapy. Modifications in the dosing regimen of TMZ
show modest improvements in progression-free survival.
Standard dosing regimens of TMZ (200 mg/m2)producea6
month progression-free survival of 21% [34]. TMZ delivered
in a low-dose, protracted schedule (75 mg/m2for 21 days of
a 28-day cycle) in an attempt to deplete MGMT produced
PFS-6 of 30% [35].
4. Targeted Therapy
Considering glioblastoma treatment is the most expensive
cancer treatment per capita in the United States, and
outcomes are still so universally poor, there is a great need for
more eective therapeutic options. Targeted therapy and per-
sonalized medicine are currently two of the more aggressively
pursued ideas in cancer treatment. While targeted therapies
aim to aect a specific alteration, most chemotherapies
are generic, broad-based DNA-damaging agents that aect
all cells in a similar manner. Targeted therapies oer the
possibility of selectively killing cancer cells and sparing
normal tissue. An example of a commonly targeted pathway
in glioblastoma is the EGFR receptor tyrosine kinase.
5. Receptor Tyrosine Kinase (RTK) Inhibitors
5.1. EGFR. As mentioned above, EGFR is known to be
an important player in gliomagenesis and in the aggressive
and therapeutic-resistant phenotype demonstrated by this
tumor. In addition to its critical role in several survival
signaling pathways, alterations in EGFR are some of the most
common mutations found in GBM. EGFR is overexpressed
in approximately 50% of tumors, and of those nearly half
express the constitutively active EGFRvIII mutant [36].
These combined facts have made this growth factor receptor
a very popular target for molecular therapies. Many clinical
trials have examined the ecacy of EGFR inhibitors, and
to date there is little evidence to support their use in a
monotherapy setting. While gefitinib, a selective inhibitor
of EGFRs tyrosine kinase domain (Figure 1), is approved
for use in non-small-cell lung cancer, a recent phase II
clinical trial of gefitinib in newly diagnosed GBM patients
by the Mayo/North Central Cancer Treatment Group showed
no significant improvement in either overall survival or
progression-free survival [37]. Another recent phase II trial
studying the ecacy of erlotinib, which also inhibits the
tyrosine kinase domain of EGFR (Figure 1), and TMZ
with RT for newly diagnosed GBM had to be stopped
4Neurology Research International
RTK (EGFR, VEGFR, IGFR,
PDGFK, HER2, etc.)
RTKi
Gefitinib (EGFR)
Erlotonib (EGFR)
Lapatinib (EGFR/HER2)
Imatinib (PDGFR)
NVP AEW541 (IGFR)
Cell growth,
proliferation, survival
Monoclonal
antibodies
bevacizumab
Cal-101, BKM120
MK-2206
Temsirolimus (CCI-779)
Everolimus (RAD001)
Dual PI3K/mTOR
PI-103
PKI-548, PKI-402
AKT
Notch re ceptor
GSIs
Oncolytic virus
CTL
Cell death
Stem cell
maintenance,
differentiation,
proliferation
Dendritic cell (APC)
P
PI3K
MHC
Class I
Delivery: toxins ,
siRNA, target genes
mTOR
Cell lysis
PARP: inhibitors (DNA repair)
HDAC: inhibitors (vorinostat)
DNA
mRNA
miRNA
Nucleus
P
IL-13Rα2
EGFR VIII
γ-secretase
Figure 1: Novel therapies in GBM. RTKs and survival signaling pathways are major drug targets in GBM. Receptors have been targeted
extracellularly by monoclonal antibodies or intracellularly at the tyrosine kinase domain. Major nodes in survival signaling pathways
(P13K, AKT, mTOR) have been the focus of intense study and drug development. More recent approaches include stem-cell targeting
(GSIs), inhibition of DNA rapair (PARP inhibitors), and targeting a host of cellular pathways through microRNA manipulation. Novel
tumor cell killing approaches are also being studied. Oncolytic virus therapy, either alone or in combination with targeted agent delivery
and immunotherapy, are being employed to eciently kill tumor cells while sparing normal tissue. RTK, receptor tyrosine kinase; EGFR,
epidermal growth factor receptor; VEGFR, vascular endothelial growth factor receptor; IGFR, insulin-like growth factor receptor 1; PDGFR,
platelet-derived growth factor receptor; mTor, mammalian target of rapamycin; GSI, gamma-secretase inhibitor; APC, antigen-presenting
cell; CTL, cytotoxic T lymphocyte, MHC class I, major histocompatibility complex I; PARP, poly(ADP-ribose) polymerase; HDAC, histone
deacetylase inhibitor.
Neurology Research International 5
short of full accrual due to the lack of benefit and the
unreasonable toxicity, including at least three treatment-
related deaths [38]. A dierent phase II trial of erlotinib in
the same setting showed less toxicity and even demonstrated
improved median survival (19.3 months versus 14.1 months)
compared to historical controls [39]. Interestingly, the first
study only escalated erlotinib dose to 150 mg/day while the
second study, which showed improved toxicities as compared
to historical controls, escalated to a maximum dose, in some
patients, of 300 mg/day. Overall results from multiple clinical
trials show monotherapy EGFR inhibition, or addition of
these inhibitors to standard-of-care treatments has shown
little benefit to patients and a dramatically increased toxicity
profile.
In terms of current multitherapy strategies, combin-
ing several RTK inhibitors, or attacking several important
glioma survival strategies, is likely to improve outcomes
over monotherapies, while also possibly reducing toxicity.
A recent study showed that EGFR expression in a GBM
xenograft model increased the ecacy of an anti-vascular
endothelial growth factor (VEGF) (vandetanib or cediranib)
therapy in combination with irradiation when compared
to xenografts lacking EGFR expression [40]. A preclinical
study of monoclonal antibody inhibition of both EGFR
and VEGFR-2 has demonstrated improved ecacy in an
orthotopic xenograft model [41]. This research was not
performed in the presence of IR and therefore may have
less relevance to glioblastoma therapy. However, it does
demonstrate the importance, as well as the complexity, of
designing combination therapies. In a similar vein, it was
recently shown that HER2 inhibition might help overcome
EGFR resistance and increase radiosensitivity in a GBM
cell model [42]. A dierent combination of EGFRvIII
inhibition with C-met inhibition showed synergy against
PTEN null/EGFRvIII positive tumors, a very aggressive
tumor population [43]. With the promise shown, even
novel inhibitors are being tested in combination. A recent
study reported that inhibition of autophagy was able to
enhance the cell-death-inducing capabilities of erlotinib in
a glioma cell model [44]. Lovastatin, a member of the
statin family (normally used to reduce cholesterol), was also
shown to increase the ecacy of EGFR inhibitor therapies
[45]. However, in a clinical phase I/II trial of lapatinib,
a dual EGFR/HER2 inhibitor, showed poor results [46].
Even rational combinations targeting this pathway have not
led to expected results, as shown by the underwhelming
results in a recent phase II trial examining erlotinib ther-
apy in combination with an mTOR inhibitor (sirolimus)
[47]. While EGFR inhibitor combination treatments have
produced improved results compared to monotherapy, we
are still a long way from being able to determine what
combinations will provide benefit for which patients.
5.2. Insulin-Like Growth Factor Receptor I (IGFR). While
EGFR has been the major focus of targeted receptor tyrosine
kinase therapies, there has been work into other known
survival signaling activating receptors such as IGFR. It has
previously been established that there is significant cross-talk
between IGFR and EGFR receptors, and the similar cellular
responses to signaling through these receptors could play
a large role in mediating resistance to anti-EGFR therapies
[48]. In 2002, Chakravarti et al. showed upregulation of the
IGFR gene in a GBM cell line resistant to AG1478, an EGFR
TKI. In this work, they demonstrated that upregulation of
IGFR in this resistant cell line correlated with sustained
activation of the PI3K pathway. Cotargeting of the IGFR
and the EGFR receptors led to increased apoptosis, as well
as a reduction in invasive potential [49]. A more recent
study demonstrated that combination of the IGFR inhibitor
NVP-AEW541 with dasatinib led to increased apoptosis in
GBM cell lines, but not in nonneoplastic human astrocytes,
and synergistically inhibited clonogenic survival [50]. These
studies highlight the possible ecacy of cotargeting IGF
and EGF receptors to overcome therapeutic resistance and
enhance therapeutic gain.
5.3. Platelet-Derived Growth Factor Receptor (PDGFR).
PDGF signaling is another commonly altered signaling path-
way in glioblastoma. A recent study in GBM cell lines showed
that varying concentration of imatinib, a PDGFR inhibitor,
had either cytostatic eects, at low concentrations, and pos-
sibly cytotoxic events at high concentrations [51]. Compar-
atively, another report in GBM cell lines showed treatment
with imatinib actually led to the activation and sustained
signaling through the ERK1/2, PI3K, and other important
cell survival signaling pathways [52]. Some reports have even
identified that PDGFR status was not predictive of imatinib
ecacy even though it was shown to be a prognostic marker
[53]. These results indicate that while PDGFR inhibition
might be an interesting target, much more study is needed.
6. PI3K/AKT/mTOR Inhibitors
In addition to EGFR and other RTK therapies, there has
been a major focus on inhibiting downstream survival
signaling pathways stimulated by these receptors. The two
most prominent and most studied pathways are the MAPK
signaling cascade and the PI3K/AKT/mTOR pathway. Ini-
tially it was believed PI3K signaling was responsible for
cell survival, while the MAPK pathway was involved in cell
proliferation. Now, these two pathways are thought to share
a significant amount of overlap and to both be involved in
cell growth, proliferation, and survival. As targeted therapies
have become a more important piece of the cancer treatment
arsenal, these pathways have been the focus of a significant
amount of research eort.
One of the initial works identifying PI3K inhibitors as
viable for the treatment of GBM was a paper by Kubota
et al. which showed the early PI3K inhibitor wortmannin
sensitized GBM cells to radiation regardless of p53 status
[54]. Wortmannin was then later shown to reverse the
growth advantage seen in GBM cells which both lacked
PTEN expression and overexpressed the EGFRvIII “always-
on” variant growth receptor [55]. However wortmannin,
while potent, has significant levels of nonspecific kinase inhi-
bition and is soluble in organic solvents, which has limited
its applicability for human clinical trials [56]. Following
the proof of principle of this novel targeting therapy, many
6Neurology Research International
new PI3K inhibitors were developed and used in clinical
trials. These include perifosine, cal101, px-866, pi-103, and
others with some PI3K inhibitors even being specifically
assessed in glioblastoma (XL765, XL147, and BKM 120)
(Figure 1). Results of many of these trials have been poor;
however, therapies using these drugs in combination with
other inhibitors have recently become a focus.
In addition to PI3K inhibition, small-molecule inhibitors
have been developed both upstream (i.e., AKT inhibitors)
and downstream (i.e., mTOR inhibitors) in this pathway.
AKT has been targeted because this kinase is the central node
in the RTK/PI3K/AKT/mTOR signaling cascade. Direct inhi-
bition of this molecule would prevent downstream signaling
similar to RTKi or PI3K inhibition. The importance of devel-
oping these novel inhibitors at dierent stages of the signal-
ing cascade has become even more obvious with the devel-
opment of RTKi refractory tumors. Only one AKT inhibitor,
mk-2206, has currently made it into phase II clinical trials.
Recently GlaxoSmithKline has begun phase I trials with
two dierent AKT inhibitors, with mixed results. The initial
phase I study of drug GSK690693 was withdrawn, and trials
of drug GSK2141795 are currently not recruiting patients.
Also, a phase II trial of MK-2206 had been planned in recur-
rent glioma; however, that trial has since been withdrawn.
However promising it has been preclinically, AKT inhibition
has proven dicult to translate into clinical ecacy.
Probably the most targeted member of this pathway is the
mammalian target of rapamycin (mTOR). In gliomas, it has
been observed that the mTORC2 complex promotes growth
and cell motility [57]. An early study demonstrating ecacy
of targeting this pathway showed increased radiosensitization
of a U87 xenograft [58]. Based on available data, it seems
likely that in order to eectively block mTOR activity in
cells, both mTORC1 and mTORC2 complexes will need to
be targeted [59]. These preclinical results have been critical
to planning the numerous clinical trials that have been
performed with mTOR inhibitors in glioblastoma. A phase
I trial of rapamycin in PTEN-deficient glioblastoma patients,
while showing some promising results, also demonstrated
the inherent diculties of targeting this protein. In this
trial, multiple patients were observed who showed elevated
levels of pAKT following mTOR inhibition, which was
correlated with shorter time to progression [60]. The AKT
activation observed was likely due to alteration of signaling
feedback loops, again highlighting the complexity of targeted
therapy. Combination therapy to block these feedback loops
may also improve ecacy [61]. Despite this complexity,
promising results have pushed mTOR inhibitors to further
trials. Several of these mTOR inhibitors have been or are
currently being tested in the clinical trials setting specifically
in gliomas, including temsirolimus, everolimus (RAD001),
and sirolimus. Temsirolimus (CCI-779) has been the most
extensively studied drug in clinical trials. A phase I study
determined the clinically eective dose to be 250 mg IV
weekly [57]. Phase II trials with CCI-779 as a monotherapy in
recurrent GBM showed no eectiveness despite low toxicity
and initial disease stabilization [62]; however, a North
Central Cancer Treatment Group study showed a statistically
significant time to progression increase in temsirolimus
responders (5.4 months versus 1.9 months, 2.3 months
overall) [63].
Because of the promise of combination therapy, cur-
rently there is a significant emphasis on dual PI3K/mTOR
inhibitors. Several novel small-molecule inhibitors have been
developed that have dual specificity for these targets. XL765
has recently been shown to reduce cell viability in vitro
and in limited animal study showed a possible eectiveness
when combined with TMZ therapy [64]. Similarly PKI-
587 and PKI-402 were shown to have a strong in vitro
antitumorigenic eect across multiple cell types including
glioma cells, while also slowing tumor growth in xenograft
models [65,66]. Another dual PI3K/mTOR inhibitor, PI-
103, which is known to have monotherapy ecacy in glioma
[67] was recently shown to specifically reduce tumor volumes
in combination with NSC-delivered s-trail in an orthotopic
intracranial xenograft model [68]. PI-103 combination ther-
apy has also proven eective in sensitizing cells to both
chemotherapy [69] and radiation [70] through reducing
DNA damage repair. RAD001 is currently being used in
multiple combination treatment studies, including studies
employing an oncolytic virus [71], Raf inhibitors [72], and
VEGFR-2 [73]. In a GBM orthotopic xenograft model, it was
shown however that PTEN does not serve as a predictive
marker for RAD001 eectiveness, despite the importance of
PI3K/AKT signaling activation in mTOR upregulation [74].
There has also been evidence for targeting this signaling
pathway from both ends, with a report showing rapamycin
promotes a response to EGFR inhibitors in either PTEN-
sucient or PTEN-deficient GBM cells by reducing tumor
cell growth. This treatment combination also results in
tumor cell death in PTEN-deficient cells [75]. While there
is hope clinical trials with these novel dual-targeting agents
will demonstrate better ecacy than current monotherapies,
there are still significant diculties in trying to determine the
best role for these targeted therapies in cancer treatment.
7. Antiangiogenic Therapies
Antiangiogenic therapy, which has been well studied in
many types of cancer, has also emerged as a novel therapy
for glioblastoma. Glioblastoma is characterized by vascu-
lar proliferation or angiogenesis [76], and advances in
molecular biology have allowed us to target angiogenesis
of glioblastoma. VEGF, a critical mediator of angiogenesis,
is highly expressed in glioblastoma and regulates tumor
angiogenesis [77,78]. Preclinical studies have shown that
VEGF inhibitors inhibit the growth of glioma cells [79,80].
Antiangiogenic therapies for glioblastoma are currently the
most advanced of any targeted therapy, and many clinical
trials have demonstrated their ecacy. In fact, bevacizumab
has been approved by the US food and Drug Administration
in the setting of recurrent glioblastoma.
Bevacizumab is a humanized monoclonal antibody
against VEGF and prevents the activation of VEGF receptor
tyrosine kinases (Figure 1)[81]. This drug is considered a
well-established antiangiogenic therapy in several angiogenic
tumors [82]. In glioblastoma, a phase II study of the addition
of bevacizumab to the standard treatment of TMZ and
Neurology Research International 7
radiotherapy was conducted for 70 newly diagnosed patients
[83]. The median overall survival and PFS were 19.6 and 13.6
months, respectively. Another phase II study of additional
bevacizumab to standard therapy showed median PFS was
13.8 months in 125 newly diagnosed glioblastoma patients
[84]. Recently two clinical trials of bevacizumab have been
reported in 2011 ASCO annual meeting for newly diagnosed
glioblastoma. Vredenburgh et al. performed a phase II
study of bevacizumab, TMZ, and radiotherapy followed by
bevacizumab, TMZ, and oral topotecan [85]. The 6-month
event-free survival was 90%, and median overall survival has
not been reached. Although the regimen was tolerable, there
were 2 treatment-related deaths, including CNS hemorrhage
and pneumonitis. Omuro et al. conducted a phase II trial
of bevacizumab, TMZ, and hypofractionated stereotactic
radiotherapy for newly diagnosed glioblastoma patients with
tumor volume under 60 cc [86]. The median PFS was 11
months, and objective response rate was 90%. Furthermore,
1-year overall survival was 90% with median follow-up of
13 months. Despite a more aggressive radiotherapy schedule,
the regimen was well tolerated and had promising results.
Additional bevacizumab seems to have favorable eects;
however, it is still unclear whether this regimen can improve
overall survival. Two randomized phase III trials, RTOG
0825 and AVAGLIO, are ongoing to demonstrate the ecacy
and safety of combined therapy of bevacizumab, TMZ, and
radiotherapy for newly diagnosed glioblastoma [87,88].
The addition of bevacizumab to TMZ and radiotherapy is
expected to become frontline treatment for glioblastoma,
and is already thought of by some as nearly a part of standard
of care.
While bevacizumab has been thoroughly investigated
in clinical and preclinical studies, other drugs have also
been studied as antiangiogenic therapy for glioblastoma
patients [89]. Cilengitide, which is selective for αvβ3and
αvβ5 integrin receptors, is considered a novel antiangiogenic
therapy for glioblastoma. A phase II study of recurrent
glioblastoma treated by cilengitide showed that 6 month PFS
was 15%, and treatment was well tolerated [90]. Furthermore
a phase I/IIa study of cilengitide combined with TMZ
and radiotherapy was performed for 52 newly diagnosed
glioblastoma patients [91]. This regimen was well tolerated
and showed promising results with median overall survival
of 16.1 months. Currently, two randomized trials, CENTRIC
and CORE, are ongoing and are expected to show the benefits
of additional cilengitide to standard therapy for newly diag-
nosed glioblastoma patients [88,92]. Other antiangiogenic
therapies (e.g., VEGF receptor tyrosine kinase inhibitors)
have also been performed in clinical and preclinical studies
[89], although there have not been any drugs to show strong
antiglioma eects compared with bevacizumab. Further
investigation is warranted to establish the ecacy and safety
of novel antiangiogenic therapy for glioblastoma.
8. Novel Targeted Therapies
8.1. Notch Inhibitors. One of the most controversial topics in
cancer biology is the theory of cancer stem cells or tumor-
initiating cells. Despite the split opinions regarding the
existence of this cell population, mounting evidence has
spurred development of novel therapeutics to target this
proposed group of cells. While proper definition and
identification of this stem cell population is still ongoing,
researchers have used clues about pathways critical to
known stem cell populations to design novel therapies.
One such pathway is the notch signaling pathway, which
is important in both normal and neoplastic development
in the central nervous system by controlling proliferation,
apoptosis, stem cell maintenance, dierentiation, and home-
ostasis [93]. Specifically in gliomas, notch has been linked
to overexpression of EGFR [94]; however, it is more likely
notch’s role in the maintenance of stem cell populations that
make it an interesting therapeutic target [95,96]. Gamma
secretase inhibitors (GSIs), which are known to inhibit
notch, have been shown to inhibit glioma stem cell growth
[97], while overexpression of notch induces tumor growth
and can be blocked by treatment with GSIs (Figure 1). While
notch inhibition is still very novel, it has shown ecacy
in preclinical models and shows a strong possibility for
combination therapy targeting both tumor cell bulk with
conventional therapies as well as the tumor-initiating cell
population.
8.2. Virotherapy/Gene Therapy for GBMs. Cancer therapy
using viruses comes in a variety of approaches including
direct viral cytotoxicity and targeted toxin delivery. The
use of viruses to deliver tumor suppressors, or siRNAs, to
knockdown oncogene expression, immune modulating com-
pounds, or antiangiogenic compounds, is also underway, yet
these attempts are often designed in combination, or as a
method to enhance oncolytic viruses (Figure 1).
The general goal behind direct viral cytotoxicity or
oncolytic virotherapy is to design a virus that specifically
and faithfully infects and replicates only in tumor cells. This
is usually accomplished by attenuating the virus to restrict
their replication to actively dividing cells (i.e., tumor cells)
while sparing normal, nonreplicating tissue. Viruses such
as herpes simplex virus 1 (HSV), adenovirus, and reovirus
have been attenuated so as to conditionally replicate within
cancer cells [98100]. Oncolytic viral therapy has undergone
major changes, using the virus not only as a cytotoxic
therapy, but also as a delivery mechanism. Researchers
have used oncolytic HSV-1 to deliver vasculostatin, an
antiangiogenic compound [101], as well as chondroitinase
ABC I [102]. This method has demonstrated significantly
enhanced therapeutic ecacy over virus alone. Similar
approaches with adenoassociated virus (aav) particles [103]
have shown codelivery of oncolytic virus with antitumor
molecules has enhanced ecacy and improved survival in
xenograft models. Each of these oncolytic viruses has also
been examined in phase I clinical trials in glioma and have
been shown to be well tolerated [104106].
Targeted toxin delivery is similar to oncolytic virus
therapy in that it is a virus-mediated cytotoxic therapy.
While oncolytic viruses are directly responsible for tumor cell
death, the strategy for this therapy is to use nonreplicating
virusparticles,suchasaav,todeliverapowerfultoxin,
8Neurology Research International
that is, Pseudomonas exotoxin (PE), specifically to cancer
cells by targeting preferentially expressed receptors. Some of
the receptors include the IL-13R variant α2, which varies
from the receptor expressed on normal brain cells [107], as
well as EGFR [108] or the EGFRvIII variant [109]. Clinical
trials have been performed with an IL-13Rα2 targeting virus
delivering cintredekin besudotox (CB). A phase III study of
this therapy compared to Gliadel Wafer administration at
first recurrence showed no survival benefit [110]. In general,
despite promising preclinical results with viral therapy for
gliomas, clinical trials resulting from this work have yet to
show any significant survival benefits. This pattern is not
uncommon in cancer therapy, yet it does point out the need
for more research. Despite the poor clinical trial results to
date, the positive data coming from viral therapy research
indicate that this form of therapy has significant future
potential.
8.3. Immunotherapy. One alternative to target gliomas with-
out aecting normal cells is by utilizing the body’s natural
defenses to kill tumor cells. There are currently two main
immunotherapy strategies being tested against gliomas:
adoptive immunotherapy and active immunotherapy.
Adoptive immunotherapy is the process of stimulating
immune cells ex vivo and then readministering them to the
patient in hope of therapeutic benefit. This can be done
either intravenously or directly into the tumor. The two
major cell types used are lymphocyte activated killer (LAK)
cells or cytotoxic T lymphocytes (CTLs). Recent reports have
identified a synergistic response between cytokine-induced
killer cells (CIKs) and TMZ [111]. Specific targeting of HER-
2 by T cells was shown to induce regression in HER-2-
positive autologous tumor xenografts as well as to target
the tumor-initiating cell (TIC) population, demonstrating a
targeted approach may prove to be more eective [111,112].
AsignicantnumberofphaseIorphaseI/IItrialshavebeen
performed using LAK cells or CTLs. These trials were largely
in the recurrent setting and demonstrated limited ecacy
combined with significant rates of toxicity for LAK cells
[113], and better tolerance and improved survival for CTLs
[113]. While responses to the adoptive therapies have shown
only limited ecacy to date, it is possible that combination
therapy or more targeted immunotherapeutic approaches
may be of use.
Active immunotherapy is similar to vaccination, the
idea being to stimulate the patient’s immune system by
using tumor-related sources of antigen (whole tumor cells,
tumor protein lysates, mRNA, synthetic peptides). These
sources of antigen can either be injected alone or coupled
to dendritic cells [113]. Dendritic cells are powerful antigen-
presenting cells, and dendritic cell therapy is designed to
increase antigen presentation by incubating tumor antigens
with these cells before injecting them back into the body
(Figure 1). This method of active tumor immunotherapy
has been the most extensively studied with widely varying
results. Some of the trials performed using this method
have shown promising results, while others demonstrated no
benefit [113]. A major problem in interpreting results from
these trials is the wide variation in protocols for everything
from acquiring cells, type of tumor antigen chosen, and
number of cells used. However, targeting this type of therapy
may lead to enhanced clinical benefit. One major trial
using patient tumor cell cultures infected with Newcastle
Disease virus followed by gamma irradiation demonstrated
significant increases in progression-free survival, 40 weeks
versus 26 weeks, as well as overall survival, 100 weeks versus
49 weeks [114]. This trial also reported increased 1-year
(91% versus 45%), 2-year (39% versus 11%), and long-term
survivors (4% versus 0%). These results are very promising;
however, the trial was nonrandomized and studied a limited
number of subjects (23 patients receiving immunotherapy
with 87 controls). Controls were also not treated using
current standard of care as this trial was performed in 2004
before the results of the Stupp trial were published. Human
cytomegalovirus (CMV) has been identified to be associated
with tumors in a significant proportion of glioblastoma
patients (50–90%) [115,116]. Currently two clinical trials are
ongoing at the Duke Brain Tumor Immunotherapy Program
attempting to utilize this knowledge. The Vaccine Therapy
in Treating Patients with Newly Diagnosed Glioblastoma
Multiforme (ATTAC) [117] and the Evaluation of Recovery
From Drug-Induced Lymphopenia Using Cytomegalovirus-
Specific T-Cell Adoptive Transfer (ERaDICATe) [118]trials
are either recruiting or in a data analysis phase, with results
likely to be reported soon.
8.4. DNA Damage. DNA damage repair is a double-edged-
sword in the cancer world. Lack of proper DNA repair
can lead to genomic instability and the generation of
cancer. However, once cancer is established, DNA repair
genes undermine many eective cancer therapeutics. Both
radiotherapy and chemotherapies are designed around a
DNA-damaging strategy designed to induce cell death and
tumor regression. In the presence of DNA repair proteins,
these therapies have reduced ecacy. By targeting DNA
repair proteins in cancer cells, we can again render them
sensitive to radiation and chemotherapies. PARP plays a role
in single stranded DNA, repair and inhibitors are currently
being tested in a number of cancer sites. If PARP is inhibited,
single-stranded nicks will not be repaired and will lead
to DNA strand breaks during replication. Double-stranded
DNA breaks are extremely toxic lesions to cells, and thereby
PARP inhibition should increase cell death in proliferating
cells (Figure 1). PARP inhibitors E7016 and AZD2281 have
been shown to radiosensitize GBM cells both in vitro and
in vivo [119,120], with an enhanced eect when combined
with heat shock protein 90 (HSP90) inhibition [121]. PARP
inhibitors have also been shown to increase the ecacy
of chemotherapies such as DNA topoisomerase I poisons,
TMZ, iriniotecan, or cilengitide [122124]. It has also been
observed that PTEN loss can negatively aect homologous
recombination, thereby increasing the ecacy of PARP
inhibition as well as other DNA-damaging modalities [125].
In addition to PARP inhibition, other targets have been
identified to utilize DNA repair as a therapeutic strategy.
Inhibition of PP2A has been shown to augment DNA-
damaging agents by inducing Plk-1 and AKT activation
and decreasing p53 expression which has led to complete
Neurology Research International 9
remission or significant tumor regression, when combined
with TMZ or doxorubicin, in a large number of tumors in a
xenograft model [126].
8.5. Autophagy. Autophagy is an evolutionarily conserved
process through which the cell is able to degrade damaged
organelles and other cell components [127]. Reports identify
autophagy to serve a dual role in cancer. Limited and
controlled autophagy can be a survival method for cancer
cells which allows them to overcome current therapies
such as chemotherapy and radiation [128130]. However,
autophagy has also been identified as a possible therapeutic
target because sustained and uncontrolled autophagy can
lead to cell death [131,132]. This process is interlinked
with several crucial cancer survival pathways including
p53 signaling and the PI3K pathway, as well as apoptotic
signaling molecules like Bif-1 [127]. Currently, attempts
are being made to control this switch and tip cells into
a pro-death state. This work is similar to the rationale
behind apoptotic research. Resistance to apoptosis is one
of the major hallmarks of many cancer types, and the
ability to regulate this cellular process would allow for
not only increased ecacy of current treatments but also
a novel target for future therapy. As with most therapies
discussed so far, combination therapy will likely play a
key role in future treatment plans. Recent studies have
demonstrated autophagy can be induced in glioma cells
by current standard-of-care therapy [130,133] and AKT
signaling plays a major role in the prosurvival eects of this
process [129,134]. This data indicates that cotargeting AKT
will be important in regulating and controlling the prodeath
role of autophagy in glioma.
8.6. HDAC Inhibitors. HDAC inhibitors have been studied
in the setting of GBM and shown positive indications in
both preclinical and clinical testing. In preclinical models,
HDACs have been shown to sensitize cells to chemotherapy
[135], to have antiproliferative activity by increasing PTEN
and AKT expression while reducing phosphorylation of the
proteins to their active forms [136], to increase apoptosis
in GBM cells through activation of the JNK pathway and
reductionintelomeraseactivity[137], and to sensitize cells
to radiation [138]. In addition, in a phase II clinical trial,
the potent HDAC inhibitor, vorinostat, was shown to have
modest single-agent ecacy and to be well tolerated [139].
8.7. MicroRNA. As microRNAs have become better studied,
the possible roles in therapeutic scenarios have increased.
Issues still remain in targeted delivery of these microRNA
constructs to tumor cells, but many possible targets have
been identified for almost all cancer types. In glioblas-
toma, some of these targets include miR-124 and miR-137,
which could target both tumor-initiating cells, by inducing
dierentiation, and normal glioblastoma cells by arresting
cell growth [140]. The miR 302–367 cluster has also been
shown to induce TIC dierentiationaswellastoreduce
the infiltrative properties of these cells [141]. A miR-21
inhibitor has been shown to sensitize the glioblastoma cell
line U251 to ionizing radiation [142]. miR-34-a has been
shown to inhibit glioblastoma growth by targeting the c-Met
and notch pathways, two well-known signaling pathways
linked to glioma pathogenesis [143]. miR-10b has been
linked to glioblastoma cell growth [144], and miR-124a has
been linked to migration and invasion [145]. This sampling
of microRNA targets linked to glioblastoma growth, survival,
TIC maintenance, and migration/invasion underscores the
possible therapeutic application of microRNAs to regulate
major pathways linked to disease severity.
9. Discussion
In this paper we have discussed RTK inhibition, angiogenesis
inhibitors, and the PI3K/Akt/mTOR inhibition in detail
due to the substantial amount of research conducted in
these areas and have also briefly discussed several very
novel areas of research including Notch inhibition, viral
and immunotherapies, and DNA repair pathways. Within
the heavily researched pathways, the overarching theme in
glioblastoma therapy is that monotherapies demonstrate
limited ecacy. Because single-agent therapies have shown
no significant benefit, it is critical to begin designing
rational combinations. Dierent RTK inhibitors combined
with PI3K/mTOR dual inhibitors or antiangiogenic agents
combined with Akt inhibition are already being examined.
It is likely that many of the novel therapies discussed in
this work will demonstrate greater ecacy when paired with
the more studied targeted therapies. Because many of these
targets are within the same signaling cascade, inhibiting
pathways horizontally rather than vertically should remove
some of the compensatory mechanisms glioblastomas use to
overcome treatment. It is also important to note that many
of the molecular biology advancements will be augmented
by advancements in current treatments. Improved tumor
border delineation or detection of microscopic disease will
enhance the ecacy of upfront surgical and radiotherapy
interventions while better methods for posttreatment image
surveillance will improve treatments in the recurrent setting.
Critically, it should be acknowledged that these therapies
will need to work in conjunction with the current standard
of care, highlighting treatments that can serve as radio-or
chemosensitizers. Glioblastoma carries a very poor progno-
sis, but with improved technology and novel, personalized,
rational, targeted therapies patient survival and quality of life
will be greatly improved.
References
[1] E. R. Laws, I. F. Parney, W. Huang et al., “Survival following
surgery and prognostic factors for recently diagnosed malig-
nant glioma: data from the glioma outcomes project,Journal
of Neurosurgery, vol. 99, no. 3, pp. 467–473, 2003.
[2] W. Stummer, U. Pichlmeier, T. Meinel, O. D. Wiestler, F.
Zanella, and H. J. Reulen, “Fluorescence-guided surgery with
5-aminolevulinic acid for resection of malignant glioma: a
randomised controlled multicentre phase III trial,Lancet
Oncology, vol. 7, no. 5, pp. 392–401, 2006.
[3] V. Vuorinen, S. Hinkka, M. Farkkila, and J. Jaaskelainen,
“Debulking or biopsy of malignant glioma in elderly
10 Neurology Research International
people—a randomised study,Acta Neurochirurgica, vol. 145,
no. 1, pp. 5–10, 2003.
[4] K. Kristiansen, S. Hagen, T. Kollevold, A. Torvik, I. Holme,
and R. Nesbakken, “Combined modality therapy of operated
astrocytomas grade III and IV. Confirmation of the value
of postoperative irradiation and lack of potentiation of
bleomycin on survival time: a prospective multicenter trial
of the scandinavian glioblastoma study group,Cancer, vol.
47, no. 4, pp. 649–652, 1981.
[5] M.D.Walker,E.AlexanderJr,W.E.Hunt,C.S.MacCarty,
M. S. Mahaley Jr, and J. Mealey Jr, “Evaluation of BCNU
and/or radiotherapy in the treatment of anaplastic gliomas.
A cooperative clinical trial,Journal of Neurosurgery, vol. 49,
no. 3, pp. 333–343, 1978.
[6] R.Stupp,W.P.Mason,M.J.vandenBentetal.,“Radiother-
apy plus concomitant and adjuvant temozolomide for glio-
blastoma,New England Journal of Medicine, vol. 352, no. 10,
pp. 987–996, 2005.
[7]R.Stupp,M.E.Hegi,W.P.Masonetal.,“Eects of radio-
therapy with concomitant and adjuvant temozolomide ver-
sus radiotherapy alone on survival in glioblastoma in a
randomised phase III study: 5-year analysis of the EORTC-
NCIC trial,The Lancet Oncology, vol. 10, no. 5, pp. 459–466,
2009.
[8] M. Esteller, J. Garcia-Foncillas, E. Andion et al., “Inactivation
of the DNA-repair gene MGMT and the clinical response
of gliomas to alkylating agents,New England Journal of
Medicine, vol. 343, no. 19, pp. 1350–1354, 2000.
[9] M. E. Hegi, A. C. Diserens, S. Godard et al., “Clinical Trial
Substantiates the Predictive Value of O-6-Methylguanine-
DNA Methyltransferase Promoter Methylation in Glioblas-
toma Patients Treated with Temozolomide,Clinical Cancer
Research, vol. 10, no. 6, pp. 1871–1874, 2004.
[10] M. E. Hegi, A. C. Diserens, T. Gorlia et al., “MGMT gene
silencing and benefit from temozolomide in glioblastoma,
New England Journal of Medicine, vol. 352, no. 10, pp. 997–
1003, 2005.
[11] L. Souhami, W. Seiferheld, D. Brachman et al., “Random-
ized comparison of stereotactic radiosurgery followed by
conventional radiotherapy with carmustine to conventional
radiotherapy with carmustine for patients with glioblastoma
multiforme: report of radiation therapy oncology group 93-
05 protocol,International Journal of Radiation Oncology
Biology Physics, vol. 60, no. 3, pp. 853–860, 2004.
[12] N. J. Laperriere, P. M. K. Leung, S. McKenzie et al., “Ran-
domized study of brachytherapy in the initial management of
patients with malignant astrocytoma,” International Journal
of Radiation Oncology Biology Physics,vol.41,no.5,pp.
1005–1011, 1998.
[13] M. R. Gilbert, M. Wang, K. D. Aldape et al., “RTOG 0525:
a randomized phase III trial comparing standard adjuvant
temozolomide (TMZ) with a dose-dense (dd) schedule in
newly diagnosed glioblastoma (GBM),Journal of Clinical
Oncology, vol. 29, no. 15, 2011.
[14] D. Pauleit, G. Stoels, A. Bachofner et al., “Comparison of
(18)F-FET and (18)F-FDG PET in brain tumors,Nuclear
Medicine and Biology, vol. 36, no. 7, pp. 779–787, 2009.
[15] I. H. Lee, M. Piert, D. Gomez-Hassan et al., “Association of
11C-methionine PET uptake with site of failure after concur-
rent temozolomide and radiation for primary glioblastoma
multiforme,” International Journal of Radiation Oncology
Biology Physics, vol. 73, no. 2, pp. 479–485, 2009.
[16] N. Kawai, Y. Maeda, N. Kudomi et al., “Correlation of bio-
logical aggressiveness assessed by 11C-methionine PET and
hypoxic burden assessed by 18F-fluoromisonidazole PET in
newly diagnosed glioblastoma,European Journal of Nuclear
Medicine and Molecular Imaging, pp. 1–10, 2010.
[17] N. Sadeghi, I. Salmon, C. Decaestecker et al., “Stereotactic
comparison among cerebral blood volume, methionine
uptake, and histopathology in brain glioma,Amer ican
Journal of Neuroradiology, vol. 28, no. 3, pp. 455–461, 2007.
[18] D. Pauleit, F. Floeth, K. Hamacher et al., “O-(2-[18F]flu-
oroethyl)-L-tyrosine PET combined with MRI improves the
diagnostic assessment of cerebral gliomas,Brain, vol. 128,
no. 3, pp. 678–687, 2005.
[19] M. D. Piroth, M. Pinkawa, R. Holy et al., “Prognostic Value of
Early [18F]Fluoroethyltyrosine Positron Emission Tomogra-
phy After Radiochemotherapy in Glioblastoma Multiforme,
International Journal of Radiation Oncology, Biology, Physics,
2010.
[20] A. P. Krishnan, I. M. Asher, D. Davis, P. Okunie,andW.
G. O’Dell, “Evidence That MR Diusion Tensor Imaging
(Tractography) Predicts the Natural History of Regional
Progression in Patients Irradiated Conformally for Primary
Brain Tumors,International Journal of Radiation Oncology
Biology Physics, vol. 71, no. 5, pp. 1553–1562, 2008.
[21] I. Yang and M. K. Aghi, “New advances that enable identi-
fication of glioblastoma recurrence,Nature Reviews Clinical
Oncology, vol. 6, no. 11, pp. 648–657, 2009.
[22] W. Hollingworth, L. S. Medina, R. E. Lenkinski et al., “A sys-
tematic literature review of magnetic resonance spectroscopy
for the characterization of brain tumors,” American Journal
of Neuroradiology, vol. 27, no. 7, pp. 1404–1411, 2006.
[23] M. Patel, F. Siddiqui, J. Y. Jin et al., “Salvage reirradiation
for recurrent glioblastoma with radiosurgery: radiographic
response and improved survival,Journal of Neuro-Oncology,
vol. 92, no. 2, pp. 185–191, 2009.
[24] P. C. Hsieh, J. P. Chandler, S. Bhangoo et al., “Adjuvant
gamma knife stereotactic radiosurgery at the time of tumor
progression potentially improves survival for patients with
glioblastoma multiforme,Neurosurgery.,vol.57,no.4,pp.
684–692, 2005.
[25] T. Biswas, P. Okunie, M. C. Schell et al., “Stereotactic radio-
surgery for glioblastoma: retrospective analysis,Radiation
Oncology, vol. 4, p. 11, 2009.
[26] S. E. Fogh, D. W. Andrews, J. Glass et al., “Hypofractionated
stereotactic radiation therapy: an eective therapy for recur-
rent high-grade gliomas,” Journal of Clinical Oncology, vol.
28, no. 18, pp. 3048–3053, 2010.
[27] E. Fokas, U. Wacker, M. W. Gross, M. Henzel, E. Encheva, and
R. Engenhart-Cabillic, “Hypofractionated stereotactic reir-
radiation of recurrent glioblastomas: a beneficial treatment
option after high-dose radiotherapy?” Strahlentherapie und
Onkologie, vol. 185, no. 4, pp. 235–240, 2009.
[28] S. E. Combs, C. Thilmann, L. Edler, J. Debus, and D. Schulz-
Ertner, “Ecacy of fractionated stereotactic reirradiation in
recurrent gliomas: long-term results in 172 patients treated
in a single institution,Journal of Clinical Oncology, vol. 23,
no. 34, pp. 8863–8869, 2005.
[29] S.F.Shepherd,R.W.Laing,V.P.Cosgroveetal.,“Hypofrac-
tionated stereotactic radiotherapy in the management of
recurrent glioma,International Journal of Radiation Oncol-
ogy Biology Physics, vol. 37, no. 2, pp. 393–398, 1997.
[30] P. H. Gutin, F. M. Iwamoto, K. Beal et al., “Safety and
Ecacy of Bevacizumab With Hypofractionated Stereotactic
Irradiation for Recurrent Malignant Gliomas,International
Neurology Research International 11
Journal of Radiation Oncology Biology Physics, vol. 75, no. 1,
pp. 156–163, 2009.
[31] J. Glass, C. L. Silverman, R. Axelrod, B. W. Corn, and D. W.
Andrews, “Fractionated stereotactic radiotherapy with cis-
platinum radiosensitization in the treatment of recurrent,
progressive, or persistent malignant astrocytoma,American
Journal of Clinical Oncology, vol. 20, no. 3, pp. 226–229, 1997.
[32] G. Lederman, M. Wronski, E. Arbit et al., “Treatment of
recurrent glioblastoma multiforme using fractionated stereo-
tactic radiosurgery and concurrent paclitaxel,American
Journal of Clinical Oncology, vol. 23, no. 2, pp. 155–159, 2000.
[33] R. E. Wurm, D. A. Kuczer, L. Schlenger et al., “Hypofraction-
ated stereotactic radiotherapy combined with topotecan in
recurrent malignant glioma,International Journal of Radia-
tion Oncology Biology Physics, vol. 66, no. 4, pp. S26–S32,
2006.
[34]W.K.A.Yung,R.E.Albright,J.Olsonetal.,“Aphase
II study of temozolemide vs. procarbazine in patients with
glioblastoma multiforme at first relapse,British Journal of
Cancer, vol. 83, no. 5, pp. 588–593, 2000.
[35] A. A. Brandes, A. Tosoni, G. Cavallo et al., “Temozolomide 3
weeks on and 1 week oas first-line therapy for recurrent
glioblastoma: phase II study from Gruppo Italiano Coop-
erativo di Neuro-Oncologia (GICNO),British Journal of
Cancer, vol. 95, no. 9, pp. 1155–1160, 2006.
[36] M. Grzmil and B. A. Hemmings, “Deregulated signalling
networks in human brain tumours,Biochimica et Biophysica
Acta—Proteins and Proteomics, vol. 1804, no. 3, pp. 476–483,
2010.
[37] J. H. Uhm, K. V. Ballman, W. Wu et al., “Phase II evaluation
of gefitinib in patients with newly diagnosed grade 4
astrocytoma: Mayo/North central cancer treatment group
study N0074,International Journal of Radiation Oncology,
Biology, Physics, 2010.
[38] D. M. Peereboom, D. R. Shepard, M. S. Ahluwalia et al.,
“Phase II trial of erlotinib with temozolomide and radiation
in patients with newly diagnosed glioblastoma multiforme,”
Journal of Neuro-Oncology, vol. 98, no. 1, pp. 93–99, 2010.
[39] M. D. Prados, S. M. Chang, N. Butowski et al., “Phase II study
of erlotinib plus temozolomide during and after radiation
therapy in patients with newly diagnosed glioblastoma
multiforme or gliosarcoma,Journal of Clinical Oncology, vol.
27, no. 4, pp. 579–584, 2009.
[40] P. R. Wachsberger, Y. R. Lawrence, Y. Liu, B. Daroczi, X.
Xu, and A. P. Dicker, “Epidermal growth factor receptor
expression modulates antitumor ecacy of vandetanib or
cediranib combined with radiotherapy in human glioblas-
toma xenografts,International Journal of Radiation Oncology
Biology Physics, vol. 82, no. 1, pp. 483–491, 2012.
[41] Y. Diao, X. H. Tian, Y. Huang, L. K. Chen, X. N. Lin, and Z. W.
Zhuang, “Enhanced cancer therapy with the combination of
EGFR and VEGFR-2 targeting in an orthotopic glioblastoma
model,Journal of Chemotherapy, vol. 22, no. 6, pp. 407–412,
2010.
[42] S. Berezowska, S. Diermeier-Daucher, G. Brockhoet al.,
“Eect of additional inhibition of human epidermal growth
factor receptor 2 with the bispecific tyrosine kinase inhibitor
AEE788 on the resistance to specific EGFR inhibition in
glioma cells,International Journal of Molecular Medicine, vol.
26, no. 5, pp. 713–721, 2010.
[43] B. Lal, C. R. Goodwin, Y. Sang et al., “EGFRvIII and c-Met
pathway inhibitors synergize against PTEN-null/EGFRvIII+
glioblastoma xenografts,Molecular Cancer Therapeutics, vol.
8, no. 7, pp. 1751–1760, 2009.
[44] B. Lal, C. R. Goodwin, Y. Sang et al., “EGFRvIII and c-Met
pathway inhibitors synergize against PTEN-null/EGFRvIII+
glioblastoma xenografts,Molecular Cancer Therapeutics, vol.
8, no. 7, pp. 1751–1760, 2009.
[45] C. Cemeus, T. T. Zhao, G. M. Barrett, I. A. Lorimer, and
J. Dimitroulakos, “Lovastatin enhances gefitinib activity in
glioblastoma cells irrespective of EGFRvIII and PTEN status,
Journal of Neuro-Oncology, vol. 90, no. 1, pp. 9–17, 2008.
[46] B. Thiessen, C. Stewart, M. Tsao et al., “A phase I/II trial
of GW572016 (lapatinib) in recurrent glioblastoma multi-
forme: clinical outcomes, pharmacokinetics and molecular
correlation,Cancer Chemotherapy and Pharmacology, vol.
65, no. 2, pp. 353–361, 2010.
[47] D. A. Reardon, A. Desjardins, J. J. Vredenburgh et al., “Phase
2 trial of erlotinib plus sirolimus in adults with recurrent
glioblastoma,Journal of Neuro-Oncology,vol.96,no.2,pp.
219–230, 2010.
[48] J. van der Veeken, S. Oliveira, R. M. Schielers, G. Storm,
P.M.P.VanBergenEnHenegouwen,andR.C.Roovers,
“Crosstalk between epidermal growth factor receptor- and
insulin-like growth factor-1 receptor signaling: implications
for cancer therapy,Current Cancer Drug Targets, vol. 9, no.
6, pp. 748–760, 2009.
[49] A. Chakravarti, J. S. Loeer, and N. J. Dyson, “Insulin-
like growth factor receptor I mediates resistance to anti-
epidermal growth factor receptor therapy in primary human
glioblastoma cells through continued activation of phospho-
inositide 3-kinase signaling,Cancer Research, vol. 62, no. 1,
pp. 200–207, 2002.
[50] D. R. Premkumar, E. P. Jane, and I. F. Pollack, “Co-admin-
istration of NVP-AEW541 and dasatinib induces mitochon-
drial- mediated apoptosis through Bax activation in malig-
nant human glioma cell lines,International Journal of
Oncology, vol. 37, no. 3, pp. 633–643, 2010.
[51] E. Ranza, G. Mazzini, A. Facoetti, and R. Nano, “In-
vitro eects of the tyrosine kinase inhibitor imatinib on
glioblastoma cell proliferation,Journal of Neuro-Oncology,
vol. 96, no. 3, pp. 349–357, 2010.
[52] Y. Dong, L. Jia, X. Wang et al., “Selective inhibition of PDGFR
by imatinib elicits the sustained activation of ERK and
downstream receptor signaling in malignant glioma cells,
International Journal of Oncology, vol. 38, no. 2, pp. 555–569,
2011.
[53] J. Paulsson, M. B. Lindh, M. Jarvius et al., “Prognostic but not
predictive role of platelet-derived growth factor receptors in
patients with recurrent glioblastoma,International Journal
of Cancer, vol. 128, no. 8, pp. 1981–1988, 2011.
[54] N. Kubota, S. Okada, T. Inada, K. Ohnishi, and T. Ohnishi,
“Wortmannin sensitizes human glioblastoma cell lines car-
rying mutant and wild type TP53 gene to radiation,Cancer
Letters, vol. 161, no. 2, pp. 141–147, 2000.
[55] M. Klingler-Homann, P. Bukczynska, and T. Tiganis, “Inhi-
bition of phosphatidylinositol 3-kinase signaling negates the
growth advantage imparted by a mutant epidermal growth
factor receptor on human glioblastoma cells,International
Journal of Cancer, vol. 105, no. 3, pp. 331–339, 2003.
[56] J. LoPiccolo, G. M. Blumenthal, W. B. Bernstein, and P. A.
Dennis, “Targeting the PI3K/Akt/mTOR pathway: eective
combinations and clinical considerations, Drug Resistance
Updates, vol. 11, no. 1-2, pp. 32–50, 2008.
[57] S. M. Chang, J. Kuhn, P. Wen et al., “Phase I/pharmacokinetic
study of CCI-779 in patients with recurrent malignant
glioma on enzyme-inducing antiepileptic drugs,” Investiga-
tional New Drugs, vol. 22, no. 4, pp. 427–435, 2004.
12 Neurology Research International
[58] J. S. Eshleman, B. L. Carlson, A. C. Mladek, B. D. Kastner, K.
L. Shide, and J. N. Sarkaria, “Inhibition of the mammalian
target of rapamycin sensitizes U87 xenografts to fractionated
radiation therapy,Cancer Research, vol. 62, no. 24, pp. 7291–
7297, 2002.
[59]N.Gulati,M.Karsy,L.Albert,R.Murali,andM.Jhanwar-
Uniyal, “Involvement of mTORC1 and mTORC2 in reg-
ulation of glioblastoma multiforme growth and motility,
International Journal of Oncology, vol. 35, no. 4, pp. 731–740,
2009.
[60] T. F. Cloughesy, K. Yoshimoto, P. Nghiemphu et al., “Antitu-
mor activity of rapamycin in a phase I trial for patients with
recurrent PTEN-deficient glioblastoma,PLoS Medicine, vol.
5, no. 1, pp. 0139–0151, 2008.
[61]L.Albert,M.Karsy,R.Murali,andM.Jhanwar-Uniyal,
“Inhibition of mTOR activates the MAPK pathway in
glioblastoma multiforme,Cancer Genomics and Proteomics,
vol. 6, no. 5, pp. 255–261, 2009.
[62] S. M. Chang, P. Wen, T. Cloughesy et al., “Phase II study
of CCI-779 in patients with recurrent glioblastoma multi-
forme,Investigational New Drugs, vol. 23, no. 4, pp. 357–361,
2005.
[63]E.Galanis,J.C.Buckner,M.J.Maureretal.,“PhaseII
trial of temsirolimus (CCI-779) in recurrent glioblastoma
multiforme: a north central cancer treatment group study,
Journal of Clinical Oncology, vol. 23, no. 23, pp. 5294–5304,
2005.
[64] G. Prasad, T. Sottero, X. Yang et al., “Inhibition of
PI3K/mTOR pathways in glioblastoma and implications for
combination therapy with temozolomide,Neuro-Oncology,
vol. 13, no. 4, pp. 384–392, 2011.
[65] R. Mallon, I. Hollander, L. Feldberg et al., “Antitumor
ecacy profile of PKI-402, a dual phosphatidylinositol 3-
kinase/Mammalian target of rapamycin inhibitor,Molecular
Cancer Therapeutics, vol. 9, no. 4, pp. 976–984, 2010.
[66] Q. W. Fan, Z. A. Knight, D. D. Goldenberg et al., “A dual PI3
kinase/mTOR inhibitor reveals emergent ecacy in glioma,
Cancer Cell, vol. 9, no. 5, pp. 341–349, 2006.
[67] Q. W. Fan, Z. A. Knight, D. D. Goldenberg et al., “A dual PI3
kinase/mTOR inhibitor reveals emergent ecacy in glioma,
Cancer Cell, vol. 9, no. 5, pp. 341–349, 2006.
[68] T. Bagci-Onder, H. Wakimoto, M. Anderegg, C. Cameron,
and K. Shah, “A dual PI3K/mTOR inhibitor, PI-103, cooper-
ates with stem cell-delivered TRAIL in experimental glioma
models,Cancer Research, vol. 71, no. 1, pp. 154–163, 2011.
[69] M. A. Westho, J. A. Kandenwein, S. Karl et al., “The pyr-
idinylfuranopyrimidine inhibitor, PI-103, chemosensitizes
glioblastoma cells for apoptosis by inhibiting DNA repair,”
Oncogene, vol. 28, no. 40, pp. 3586–3596, 2009.
[70] R. Prevo, E. Deutsch, O. Sampson et al., “Class I PI3 kinase
inhibition by the pyridinylfuranopyrimidine inhibitor PI-
103 enhances tumor radiosensitivity,Cancer Research, vol.
68, no. 14, pp. 5915–5923, 2008.
[71] M. M. Alonso, H. Jiang, T. Yokoyama et al., “Delta-24-RGD
in combination with RAD001 induces enhanced anti-glioma
eect via autophagic cell death,Molecular Therapy, vol. 16,
no. 3, pp. 487–493, 2008.
[72] A. B. Hjelmeland, K. P. Lattimore, B. E. Fee et al., “The
combination of novel low molecular weight inhibitors of
RAF (LBT613) and target of rapamycin (RAD001) decreases
glioma proliferation and invasion,Molecular Cancer Thera-
peutics, vol. 6, no. 9, pp. 2449–2457, 2007.
[73] R. K. Goudar, Q. Shi, M. D. Hjelmeland et al., “Combination
therapy of inhibitors of epidermal growth factor recep-
tor/vascular endothelial growth factor receptor 2 (AEE788)
and the mammalian target of rapamycin (RAD001) oers
improved glioblastoma tumor growth inhibition,Molecular
Cancer Therapeutics, vol. 4, no. 1, pp. 101–112, 2005.
[74] L. Yang, M. J. Clarke, B. L. Carlson et al., “PTEN loss does not
predict for response to RAD001 (everolimus) in a glioblasto-
ma orthotopic xenograft test panel,Clinical Cancer Research,
vol. 14, no. 12, pp. 3993–4001, 2008.
[75] M. Y. Wang, K. V. Lu, S. Zhu et al., “Mammalian target
of rapamycin inhibition promotes response to epidermal
growth factor receptor kinase inhibitors in PTEN-deficient
and PTEN-intact glioblastoma cells,Cancer Research, vol.
66, no. 16, pp. 7864–7869, 2006.
[76] J.C.Reijneveld,E.E.Voest,andM.J.B.Taphoorn,“Angi-
ogenesis in malignant primary and metastatic brain tumors,”
Journal of Neurology, vol. 247, no. 8, pp. 597–608, 2000.
[77] N. Ferrara, H. P. Gerber, and J. LeCouter, “The biology of
VEGF and its receptors,Nature Medicine,vol.9,no.6,pp.
669–676, 2003.
[78] I. H. Chaudhry, D. G. O’Donovan, P. E. C. Brenchley, H.
Reid, and I. S. D. Roberts, “Vascular endothelial growth factor
expression correlates with tumour grade and vascularity in
gliomas,Histopathology, vol. 39, no. 4, pp. 409–415, 2001.
[79] D. F. Stefanik, W. K. Fellows, L. R. Rizkalla et al., “Monoclonal
antibodies to vascular endothelial growth factor (VEGF) and
the VEGF receptor, FLT-1, inhibit the growth of C6 glioma in
amousexenograt,Journal of Neuro-Oncology, vol. 55, no. 2,
pp. 91–100, 2001.
[80] S. Bao, Q. Wu, S. Sathornsumetee et al., “Stem cell-like
glioma cells promote tumor angiogenesis through vascular
endothelial growth factor,Cancer Research, vol. 66, no. 16,
pp. 7843–7848, 2006.
[81] L. G. Presta, H. Chen, S. J. O’Connor et al., “Humanization
of an anti-vascular endothelial growth factor monoclonal
antibody for the therapy of solid tumors and other disorders,
Cancer Research, vol. 57, no. 20, pp. 4593–4599, 1997.
[82] F. Kazazi-Hyseni, J. H. Beijnen, and J. H. M. Schellens,
“Bevacizumab,Oncologist, vol. 15, no. 8, pp. 819–825, 2010.
[83] J. J. Vredenburgh, A. Desjardins, J. P. Kirkpatrick et al., “Addi-
tion of Bevacizumab to Standard Radiation Therapy and
Daily Temozolomide Is Associated with Minimal Toxicity in
Newly Diagnosed Glioblastoma Multiforme,International
Journal of Radiation Oncology, Biology, Physics, 2010.
[84] J. J. Vredenburgh, A. Desjardins, J. P. Kirkpatrick et al.,
Addition of bevacizumab to standard radiation therapy
and daily temozolomide is associated with minimal toxicity
in newly diagnosed glioblastoma multiforme,International
Journal of Radiation Oncology Biology Physics, vol. 82, no. 1,
pp. 58–66, 2012.
[85] J. J. Vredenburgh, A. Desjardins, D. A. Reardon et al., “Beva-
cizumab, temozolomide, and radiation therapy followed
by bevacizumab, temozolomide, and oral topotecan for
newly-diagnosed glioblastoma multiforme (GBM),Journal
of Clinical Oncology, vol. 13, supplement 3, pp. iii858–iii91,
2011.
[86] A. M. P. Omuro, K. Beal, and S. Karimi, “Phase II study of
bevacizumab (BEV), temozolomide (TMZ), and hypofrac-
tionated stereotactic radiotherapy (HFSRT) for newly diag-
nosed glioblastoma (GBM),Journal of Clinical Oncology, vol.
13, supplement 3, pp. iii41–iii68, 2011.
[87] A study of avastin (bevacizumab) in combination with
temozolomide and radiotherapy in patients with newly
Neurology Research International 13
diagnosed glioblastoma, 2010, http://clinicaltrials.gov/ct2/
show/NCT00943826.
[88] Temozolomide and radiation therapy with or without bevac-
izumab in treating patients with newly diagnosed glioblasto-
ma, 2011, http://clinicaltrials.gov/ct2/show/NCT00884741?
term=rtog+0825&rank=1.
[89] K. Beal, L. E. Abrey, and P. H. Gutin, “Antiangiogenic agents
in the treatment of recurrent or newly diagnosed glioblas-
toma: analysis of single-agent and combined modality
approaches,Radiation Oncology, vol. 6, no. 1, 2011.
[90] D. A. Reardon, K. L. Fink, T. Mikkelsen et al., “Randomized
phase II study of cilengitide, an integrin-targeting arginine-
glycine-aspartic acid peptide, in recurrent glioblastoma
multiforme,” Journal of Clinical Oncology, vol. 26, no. 34, pp.
5610–5617, 2008.
[91] R. Stupp, M. E. Hegi, B. Neyns et al., “Phase I/IIa study of
cilengitide and temozolomide with concomitant radiother-
apy followed by cilengitide and temozolomide maintenance
therapy in patients with newly diagnosed glioblastoma,”
Journal of Clinical Oncology, vol. 28, no. 16, pp. 2712–2718,
2010.
[92] Cilengitide, temozolomide, and radiation therapy in treating
patients with newly diagnosed glioblastoma and methylated
gene promoter status (CENTRIC), 2011, http://clinicaltri-
als.gov/ct2/show/NCT00689221?term=centric&rank=1.
[93] M. M. Lino, A. Merlo, and J. Boulay, “Notch signaling in
glioblastoma: a developmental drug target?” BMC Medicine,
vol. 8, 2010.
[94] C. Brennan, H. Momota, D. Hambardzumyan et al., “Glio-
blastoma subclasses can be defined by activity among signal
transduction pathways and associated genomic alterations,
PLoS ONE, vol. 4, no. 11, Article ID e7752, 2009.
[95] X. Fan, L. Khaki, T. S. Zhu et al., “NOTCH pathway block-
ade depletes CD133-positive glioblastoma cells and inhibits
growth of tumor neurospheres and xenografts,Stem Cells,
vol. 28, no. 1, pp. 5–16, 2010.
[96] M. Ying, S. Wang, Y. Sang et al., “Regulation of glioblastoma
stem cells by retinoic acid: role for Notch pathway inhibi-
tion,Oncogene, vol. 30, no. 31, pp. 3454–3467, 2011.
[97] J. Chen, S. Kesari, C. Rooney et al., “Inhibition of Notch
signaling blocks growth of glioblastoma cell lines and tumor
neurospheres,Genes and Cancer, vol. 1, no. 8, pp. 822–835,
2010.
[98]I.V.Ulasov,M.A.Tyler,A.A.Rivera,D.M.Nettlebeck,J.
T. Douglas, and M. S. Lesniak, “Evaluation of E1A double
mutant oncolytic adenovectors in anti-glioma gene therapy,
Journal of Medical Virology, vol. 80, no. 9, pp. 1595–1603,
2008.
[99] R.Chambers,G.Y.Gillespie,L.Soroceanuetal.,“Compar-
ison of genetically engineered herpes simplex viruses for the
treatment of brain tumors in a scid mouse model of human
malignant glioma,Proceedings of the National Academy of
Sciences of the United States of America,vol.92,no.5,pp.
1411–1415, 1995.
[100] M. E. Wilcox, W. Yang, D. Senger et al., “Reovirus as an onco-
lytic agent against experimental human malignant gliomas,”
Journal of the National Cancer Institute, vol. 93, no. 12, pp.
903–912, 2001.
[101] J. Hardcastle, K. Kurozumi, N. Dmitrieva et al., “Enhanced
antitumor ecacy of vasculostatin (Vstat120) expressing
oncolytic HSV-1,Molecular Therapy, vol. 18, no. 2, pp. 285–
294, 2010.
[102] N. Dmitrieva, L. Yu, M. Viapiano et al., “Chondroitinase
ABC I-mediated enhancement of oncolytic virus spread and
antitumor ecacy,Clinical Cancer Research, vol. 17, no. 6,
pp. 1362–1372, 2011.
[103] D. H. Meijer, C. A. Maguire, S. G. Leroy, and M. Sena-
Esteves, “Controlling brain tumor growth by intraventricular
administration of an AAV vector encoding IFN-β,” Cancer
Gene Therapy, vol. 16, no. 8, pp. 664–671, 2009.
[104] J. M. Markert, M. D. Medlock, S. D. Rabkin et al., “Con-
ditionally replicating herpes simplex virus mutant G207 for
the treatment of malignant glioma: results of a phase I trial,”
Gene Therapy, vol. 7, no. 10, pp. 867–874, 2000.
[105] E. A. Chiocca, K. M. Abbed, S. Tatter et al., “A phase I open-
label, dose-escalation, multi-institutional trial of injection
with an E1B-attenuated adenovirus, ONYX-015, into the
peritumoral region of recurrent malignant gliomas, in the
adjuvant setting,Molecular Therapy, vol. 10, no. 5, pp. 958–
966, 2004.
[106] P. Forsyth, G. Rold´
an,D.Georgeetal.,“AphaseItrial
of intratumoral administration of reovirus in patients
with histologically confirmed recurrent malignant gliomas,
Molecular Therapy, vol. 16, no. 3, pp. 627–632, 2008.
[107] J. Wykosky, D. M. Gibo, C. Stanton, and W. Debinski,
“Interleukin-13 receptor α2, EphA2, and Fos-related antigen
1 as molecular denominators of high-grade astrocytomas and
specific targets for combinatorial therapy,Clinical Cancer
Research, vol. 14, no. 1, pp. 199–208, 2008.
[108] H. Horita, J. Thorburn, A. E. Frankel, and A. Thorburn,
“EGFR-targeted diphtheria toxin stimulates TRAIL killing
of glioblastoma cells by depleting anti-apoptotic proteins,
Journal of Neuro-Oncology, vol. 95, no. 2, pp. 175–184, 2009.
[109] P. Grandi, J. Fernandez, O. Szentirmai et al., “Targeting HSV-
1 virions for specific binding to epidermal growth factor
receptor-vIII-bearing tumor cells,Cancer Gene Therapy, vol.
17, no. 9, pp. 655–663, 2010.
[110] S. Kunwar, S. Chang, M. Westphal et al., “Phase III random-
ized trial of CED of IL13-PE38QQR vs Gliadel wafers for
recurrent glioblastoma,Neuro-Oncology,vol.12,no.8,pp.
871–881, 2010.
[111] J. Jin, K. M. Joo, S. J. Lee et al., “Synergistic therapeutic eects
of cytokine-induced killer cells and temozolomide against
glioblastoma,Oncology Reports, vol. 25, no. 1, pp. 33–39,
2011.
[112] N. Ahmed, V. S. Salsman, Y. Kew et al., “HER2-specific
T cells target primary glioblastoma stem cells and induce
regression of autologous experimental tumors,Clinical
Cancer Research, vol. 16, no. 2, pp. 474–485, 2010.
[113] E. Vauleon, T. Avril, B. Collet, J. Mosser, and V. Quillien,
“Overview of cellular immunotherapy for patients with
glioblastoma,Clinical and Developmental Immunology, vol.
2010, Article ID 689171, 2010.
[114] H. H. Steiner, M. M. Bonsanto, P. Beckhove et al., “Antitumor
vaccination of patients with glioblastoma multiforme: a pilot
study to assess feasibility, safety, and clinical benefits,Journal
of Clinical Oncology, vol. 22, no. 21, pp. 4272–4281, 2004.
[115] K. G. Lucas, L. Bao, R. Bruggeman, K. Dunham, and C.
Specht, “The detection of CMV pp65 and IE1 in glioblastoma
multiforme,” Journal of Neuro-Oncology, vol. 103, no. 2, pp.
231–238, 2011.
[116] D. A. Mitchell, W. Xie, R. Schmittling et al., “Sensitive detec-
tion of human cytomegalovirus in tumors and peripheral
blood of patients diagnosed with glioblastoma,Neuro-
Oncology, vol. 10, no. 1, pp. 10–18, 2008.
14 Neurology Research International
[117] Vaccine therapy in treating patients with newly diag-
nosed glioblastoma multiforme (ATTAC), 2011, http://www
.clinicaltrials.gov/ct2/show/NCT00639639?term=ATTAC&
rank=1.
[118] Evaluation of recovery from drug-induced lymphope-
nia using cytomegalovirus-specific T-cell adoptive transfer
(ERaDICATe), 2011, http://www.clinicaltrials.gov/ct2/show/
NCT00693095?term=ERaDICATe&rank=1.
[119] A. L. Russo, H. C. Kwon, W. E. Burgan et al., “In vitro and
in vivo radiosensitization of glioblastoma cells by the poly
(ADP-ribose) polymerase inhibitor E7016, Clinical Cancer
Research, vol. 15, no. 2, pp. 607–612, 2009.
[120] F. A. Dungey, D. A. Loser, and A. J. Chalmers, “Replication-
dependent radiosensitization of human glioma cells by
inhibition of poly(ADP-Ribose) polymerase: mechanisms
and therapeutic potential,International Journal of Radiation
Oncology Biology Physics, vol. 72, no. 4, pp. 1188–1197, 2008.
[121] F. A. Dungey, K. W. Caldecott, and A. J. Chalmers, “Enhanced
radiosensitization of human glioma cells by combining
inhibition of poly(ADP-ribose) polymerase with inhibition
of heat shock protein 90,Molecular Cancer Therapeutics, vol.
8, no. 8, pp. 2243–2254, 2009.
[122] G. Cimmino, S. Pepe, G. Laus et al., “Poly(ADPR)polym-
erase-1 signalling of the DNA damage induced by DNA
topoisomerase I poison in D54p53wt and U251p53mut
glioblastoma cell lines,Pharmacological Research, vol. 55, no.
1, pp. 49–56, 2007.
[123] L. Tentori, C. Leonetti, M. Scarsella et al., “Systemic Admin-
istration of GPI 15427, a Novel Poly(ADP-Ribose) Polymer-
ase-1 Inhibitor, Increases the Antitumor Activity of Temo-
zolomide against Intracranial Melanoma, Glioma, Lym-
phoma,Clinical Cancer Research, vol. 9, no. 14, pp. 5370–
5379, 2003.
[124] S. J. Miknyoczki, S. Jones-Bolin, S. Pritchard et al., “Chem-
opotentiation of temozolomide, irinotecan, and cisplatin
activity by CEP-6800, a poly(ADP-ribose) polymerase
inhibitor,Molecular cancer therapeutics, vol. 2, no. 4, pp.
371–382, 2003.
[125] B. McEllin, C. V. Camacho, B. Mukherjee et al., “PTEN loss
compromises homologous recombination repair in astro-
cytes: implications for glioblastoma therapy with temozolo-
mide or poly(ADP-Ribose) polymerase inhibitors,Cancer
Research, vol. 70, no. 13, pp. 5457–5464, 2010.
[126] J. Lu, J. S. Kovach, F. Johnson et al., “Inhibition of serine/
threonine phosphatase PP2A enhances cancer chemotherapy
by blocking DNA damage induced defense mechanisms,
Proceedings of the National Academy of Sciences of the United
States of America, vol. 106, no. 28, pp. 11697–11702, 2009.
[127] C. H. Eng and R. T. Abraham, “The autophagy conundrum in
cancer: influence of tumorigenic metabolic reprogramming,
Oncogene, vol. 30, no. 47, pp. 4687–4696, 2011.
[128] W. Zhuang, B. Li, L. Long, L. Chen, Q. Huang, and Z. Q.
Liang, “Knockdown of the DNA-dependent protein kinase
catalytic subunit radiosensitizes glioma-initiating cells by
inducing autophagy,Brain Research, 2010.
[129] Q. W. Fan, C. Cheng, C. Hackett et al., “Akt and autophagy
cooperate to promote survival of drug-resistant glioma,
Science Signaling, vol. 3, no. 147, Article ID ra81, 2010.
[130] S. L. Lomonaco, S. Finniss, C. Xiang et al., “The induction of
autophagy by γ-radiation contributes to the radioresistance
of glioma stem cells,International Journal of Cancer, vol. 125,
no. 3, pp. 717–722, 2009.
[131] E. C. Filippi-Chiela, E. S. Villodre, L. L. Zamin, and G. Lenz,
Autophagy interplay with apoptosis and cell cycle regulation
in the growth inhibiting eect of resveratrol in glioma cells,
PLoS ONE, vol. 6, no. 6, Article ID e20849, 2011.
[132] W. T. Liu, C. H. Lin, M. Hsiao, and P. W. Gean, “Minocycline
inhibits the growth of glioma by inducing autophagy,
Autophagy, vol. 7, no. 2, pp. 166–175, 2011.
[133] M. Natsumeda, H. Aoki, H. Miyahara et al., “Induction
of autophagy in temozolomide treated malignant gliomas,
Neuropathology, vol. 31, no. 5, pp. 486–493, 2011.
[134] Q. W. Fan and W. A. Weiss, “Autophagy and Akt promote
survival in glioma,” Autophagy, vol. 7, no. 5, pp. 536–538,
2011.
[135] A. Bangert, S. Hacker, S. Cristofanon, K. M. Debatin, and
S. Fulda, “Chemosensitization of glioblastoma cells by the
histone deacetylase inhibitor MS275,Anti-Cancer Drugs,
vol. 22, no. 6, pp. 494–499, 2011.
[136] W. J. Huang, C. W. Lin, C. Y. Lee et al., “NBM-HD-3, a novel
histone deacetylase inhibitor with anticancer activity through
modulation of PTEN and AKT in brain cancer cells, Journal
of Ethnopharmacology, vol. 136, no. 1, pp. 156–167, 2011.
[137] V. Sharma, N. Koul, C. Joseph, D. Dixit, S. Ghosh, and E. Sen,
“HDAC inhibitor, scriptaid, induces glioma cell apoptosis
through JNK activation and inhibits telomerase activity,
Journal of Cellular and Molecular Medicine, vol. 14, no. 8, pp.
2151–2161, 2010.
[138] J. H. Kim, J. H. Shin, and I. H. Kim, “Susceptibility and radio-
sensitization of human glioblastoma cells to trichostatin
A, a histone deacetylase inhibitor,International Journal of
Radiation Oncology Biology Physics, vol. 59, no. 4, pp. 1174–
1180, 2004.
[139] E. Galanis, K. A. Jaeckle, M. J. Maurer et al., “Phase II trial
of vorinostat in recurrent glioblastoma multiforme: a north
central cancer treatment group study,Journal of Clinical
Oncology, vol. 27, no. 12, pp. 2052–2058, 2009.
[140] J. Silber, D. A. Lim, C. Petritsch et al., “miR-124 and miR-
137 inhibit proliferation of glioblastoma multiforme cells
and induce dierentiation of brain tumor stem cells,BMC
Medicine, vol. 6, p. 14, 2008.
[141] M. Fareh, L. Turchi, V. Virolle et al., “The miR 302-367
cluster drastically aects self-renewal and infiltration proper-
ties of glioma-initiating cells through CXCR4 repression and
consequent disruption of the SHH-GLI-NANOG network,
Cell Death & Dierentiation, vol. 2, no. 19, pp. 232–244, 2012.
[142] Y. Li, S. Zhao, Y. Zhen et al., “A miR-21 inhibitor enhances
apoptosis and reduces G2-M accumulation induced by
ionizing radiation in human glioblastoma U251 cells,Brain
Tumor Pathology, vol. 28, no. 3, pp. 209–214, 2011.
[143] Y. Li, F. Guessous, Z. Ying et al., “MicroRNA-34a inhibits
glioblastoma growth by targeting multiple oncogenes,Can-
cer Research, vol. 69, no. 19, pp. 7569–7576, 2009.
[144] G. Gabriely, M. Yi, R. S. Narayan et al., “Human glioma
growth is controlled by microRNA-10b,Cancer Research,
vol. 71, no. 10, pp. 3563–3572, 2011.
[145] A. Fowler, D. Thomson, K. Giles et al., “miR-124a is fre-
quently down-regulated in glioblastoma and is involved in
migration and invasion,European Journal of Cancer, 2011.
... Glioblastoma is one of the most aggressive and fatal cancer types in the central nervous system [1]. They are characterized by the rapid growth rate, high invasion capacity, and resistance to treatment. ...
... Thymoquinone (TQ) is the most active ingredient of Nigella sativa. It has a preventive effect on pathologies, such as inflammation, oxidative stress, and hypertension [1,5,6]. In addition, it inhibits cancer cells by apoptotic and autophagic pathways, while its toxicity to healthy cells is very low compared to cancer cells [7]. ...
Article
Full-text available
Glioblastoma, as an invasive tumor, is one of the most common primary malignant brain tumors. Despite maximum aggressive treatment, patients with glioblastoma have a dismal prognosis. Thymoquinone (TQ) has been found to show anti-cancer effects on different types of cancer. There are a few in vitro studies on the effect of TQ on glial tumors. However, the molecular mechanism of TQ's anti-cancer effect has not been fully elucidated. In the present study, we aimed to investigate the genotoxic, apoptotic, and cytotoxic effects of TQ on C6 rat glioma cells. C6 glioma cells were analyzed after 24 h of exposure to different concentrations of TQ by the ATP cell viability assay for cytotoxicity, comet assay for genotoxicity, 2′,7′dichlorodihydrofluorescein diacetate (H2DCF-DA) for intracellular reactive oxygen species (iROS) generation, 3.3′dihexyloxacarbocyanine iodide (DiOC6(3)) for mitochondrial membrane potential, GSH/GSSG-Glo Assay for glutathione level and Fura-2AM for intracellular calcium levels. Apoptosis induction was studied by acridine orange/ethidium bromide double staining, flow cytometry, and western blotting analyses. Caspase-3, Caspase-9, Bax, Bcl-2, and pSTAT3 protein levels were determined by the western blotting method. Cytotoxicity was enhanced by TQ in C6 glioma cells in a concentration-dependent manner. TQ also induced DNA damage, apoptosis, and increased iROS. Also, MMP and GSH levels were decreased by TQ. It inhibited pSTAT3, resulting in apoptosis induction through the regulation of anti-apoptotic and pro-apoptotic proteins. Our results suggest that TQ would be an effective treatment in glioma. Further studies should support these findings.
... The prognosis for patients with malignant gliomas is heavily reliant on the morphological and molecular genetic traits of the tumor. Glioma surgery aims to maximize tumor resection while minimizing damage to critical functional regions of the brain [106][107][108][109]. However, conventional tumor extraction procedures using white light microscopy achieve maximal resection in only 23%-50% of cases [110,111]. ...
... GBM causes mitochondrial damage resulting in a lack of energy production by oxidative phosphorylation. GBM occurrence was reported for more than 75% of all CNS tumors (Perry et al., 2012). In India, compared to other CNS tumors, GBM is the most common tumor (38%) followed by oligodendrogliomas (24.5%) (S. . ...
Article
Glioblastoma (GBM) is an aggressive malignant type of brain tumor. Targeting one single intracellular pathway might not alleviate the disease, rather it activates the other molecular pathways that lead to the worsening of the disease condition. Therefore, in this study, we attempted to target both isocitrate dehydrogenase 1 (IDH1) and IDH2, which are one of the most commonly mutated proteins in GBM and other cancer types. Here, standard precision and extra precision docking, IFD, MM-GBSA, QikProp, and molecular dynamics (MD) simulation were performed to identify the potential dual inhibitor for IDH1 and IDH2 from the enamine database containing 59,161 ligands. Upon docking the ligands with IDH1 (PDB: 6VEI) and IDH2 (PDB: 6VFZ), the top eight ligands were selected, based on the XP Glide score. These ligands produced favourable MMGBSA scores and ADME characteristics. Finally, the top four ligands 12953, 44825, 51295, and 53210 were subjected to MD analysis. Interestingly, 53210 showed maximum interaction with Gln 277 for 99% in IDH1 and Gln 316 for 100% in IDH2, which are the crucial amino acids for the inhibitory function of IDH1 and IDH2 to target GBM. Therefore, the present study attempts to identify the novel molecules which could possess a pan-inhibitory action on both IDH1 and IDH that could be crucial in the management of GBM. Yet further evaluation involving in vitro and in vivo studies is warranted to support the data in our current study.Communicated by Ramaswamy H. Sarma.
... Maximal safe resection still represents a fundamental cornerstone in glioblastoma therapy (30). Thereby, the ultimate goal constitutes the 'gross total resection,' where the contrastenhancing component of the tumor is removed without compromising neurological functions (61). Further, surgical resection allows the histopathological analysis and may normalize increased intracranial pressure or recover neuronal functions depending on the tumor location (30,62). ...
Thesis
Glioblastoma (GBM), presenting the most malignant tumor of the central nervous system, is characterized by its dynamic adaptability to microenvironmental niches. This cellular plasticity aggravates the development of an immediate resistance during tumor treatment regimens. Novel insights into the developmental trajectory exhibited by GBM displayed the remarkable capacity to respond to microenvironmental influences by clonal selection of specific phenotypes. Using the exact mechanisms, malignant GBM may develop intrinsic mechanisms to resist chemotherapeutic treatments. This resistance and other malignant hallmarks were suspected to be sustained by the paracrine and autocrine glutamate signaling via ionotropic and metabotropic receptors. This study explored the manifold concept of glutamate signaling in GBM and elucidated the regulatory role and interactions of specific glutamate receptors within the GBM microenvironment. In addition, novel insights into glutamate release dynamics are presented, underlining its function to promote tumor growth, viability, and migration in glioblastoma. By integrating diversified molecular-biological analyses, novel sequencing data analysis, and clinical data, we refined the understanding of how glutamate signaling modulates mechanisms of resistance in GBM. We characterized the functions of different ionotropic and metabotropic glutamate receptors, of which the mGluR3 receptor is highlighted by its modulatory role in supporting the capacity of GBM cells to evade standard alkylating chemotherapeutics like temozolomide. We addressed the clinical relevance of mGluR3 receptor expression in GBM patients and provided proof of concept where intrinsic mechanisms of chemoresistance and the transcriptional landscape in GBM were manipulated through the inhibition of mGluR3 to increase susceptibility to chemotherapy. Finally, we validated our findings in our human organotypic section model, where GBM growth and proliferation were significantly reduced when mGluR3 inhibition was combined with temozolomide. These findings present a new picture of how glutamate signaling via mGluR3 interacts with the phenotypical GBM transcriptional programs in the light of recently published GBM cell-state discoveries. Ultimately, our results demonstrated how glutamate signaling could be used therapeutically to overcome chemoresistance in glioblastoma. Therefore, glutamate receptors such as mGluR3 may present promising new therapeutic targets, particularly in patients with tumor recurrence or therapy resistance.
... Fortunately, brain tumors have more alkaline pH compared to healthy tissue, which helps with TMZ activation within tumor tissue, increasing its effectiveness in brain tumor therapy [173,174]. Beneficial results and improved progression-free survival in cases of recurrent glioblastoma were presented recently for bevacizumab (Avastin), a humanized monoclonal antibody that binds to vascular endothelial growth factor (VEGF) ligand, inhibiting further activation of its receptors [175]. It is noteworthy that the combination treatment of bevacizumab with radiation and TMZ for therapy of newly diagnosed glioblastoma shows some promising results, but is also associated with higher rates of toxicity [87,176,177]. ...
Article
Full-text available
The evaluation of nanomechanical properties of tissues in health and disease is of increasing interest to scientists. It has been confirmed that these properties, determined in part by the composition of the extracellular matrix, significantly affect tissue physiology and the biological behavior of cells, mainly in terms of their adhesion, mobility, or ability to mutate. Importantly, pathophysiological changes that determine disease development within the tissue usually result in significant changes in tissue mechanics that might potentially affect the drug efficacy, which is important from the perspective of development of new therapeutics, since most of the currently used in vitro experimental models for drug testing do not account for these properties. Here, we provide a summary of the current understanding of how the mechanical properties of brain tissue change in pathological conditions, and how the activity of the therapeutic agents is linked to this mechanical state.
... It is composed of N-acetyl-D-glucosamine and b-(1,4)-linked D-glucosamine linked by glycosidic bonds. It is polycationic in nature, which allows binding with negatively charged therapeutics, such as miRNA [31,32]. This polycationic nature of CS-NPs will stabilize the genetic material protect it from degradation, and promote its uptake into target cells [31]. ...
Article
Full-text available
Recent evidence has implicated microRNA-219 (miR-219) in regulation of gene contributed in glioblastoma (GBM) pathogenesis. This study aimed to prepare miR-219 in chitosan (CS) nanoparticles (NPs), characterize and investigate their efficacy on human GBM cell line (U87 MG). NPs were prepared using ionic gelation method. The influence of process parameters on physicochemical characteristics of NPs was investigated. Apoptotic effect of miR-219 was examined on U87 MG cells. Formulated NPs showed particle size of 109 ± 2.18 nm, with poly dispersity index equal to 0.2 ± 0.05, and zeta potential of +20.5 ± 0.7 mV. Entrapment efficiency of miR-219 in loaded NP has reached 95%. The in vitro release study demonstrated sustained release pattern of miR-219 from CS-NPs. Gel retardation assay has confirmed the integrity of miR-219 after production process. The fabricated NPs reduced the survival of U87 MG cells to 78% after 24 h of post-transfection, and into 67.5% after 48 h. However, fibroblasts were not affected by the NPs, revealing their specificity for GBM cells. Given the tumour suppressing function of miR-219, and advantage of CS-NPs for gene delivery to the central nervous system, the presented NPs have a great potential for treatment of GBM.
... However, advanced age, and the anatomic structures invaded by the tumour could be an impending factor for maximal surgical resection. Recent advances in technology has helped optimize safe maximal surgical resection using improved surgical imaging techniques such as intraoperative magnetic resonance imaging (iMRI) and fluorescent-guided resection (Anton et al., 2012;Perry et al., 2012;Wilson et al., 2014). Fluorescent-guided resection uses a pharmacological agent that fluoresces only when exposed to tumour cell but not the surrounding normal brain, thus helped guide tumour resection by identifying tumour tissue that may otherwise appear normal (Anton et al., 2012;Pichlmeier et al., 2008). ...
Thesis
Glioblastoma are the most aggressive adult brain tumours for which new therapeutic strategies are highly needed. They almost always recur due to resistance to radio-and chemotherapies. We are interested in integrins, in particular α5β1 integrin, as therapeutic targets in glioblastoma. Previous works showed that it participates to the temozolomide (TMZ) chemotherapy resistance by partly inhibiting the tumour suppressor p53 pathway. Here we investigated a new therapeutic option based on the combination of p53 activators and α5β1 integrin inhibitors in naïve U87MG glioma cells and their TMZ-resistant counterparts. We showed that resistance to TMZ is accompanied by a huge rearrangement of integrin expressions and that α5β1 integrin remains a pertinent target for TMZ-resistant glioblastoma. In addition, TMZ-resistant cells proved sensitive to different blockers of p53/mdm2 complexes able to reactivate the p53 signalling pathways. Our data support the hypothesis that adding p53 activators together with integrin antagonists may represent a pertinent therapeutic strategy for recurrent glioma tumours.
... Compared to Zika virus infection of differentiated GBM cells, the Zika virus preferentially infects and selectively kills GSCs and stem-like cells in a SOX2-dependent manner [220]. It has also been reported that an immunotherapeutic approach in gliomas that uses a vesicular stomatitis virus expressing HIF-2a, SOX10 and c-Myc together with checkpoint inhibitors anti-PD1 and anti-CTLA-4 enhances the antitumor response [221]. ...
Article
Full-text available
Glioblastoma (GBM) is the most common, most aggressive and deadliest brain tumor. Recently, remarkable progress has been made towards understanding the cellular and molecular biology of gliomas. GBM tumor initiation, progression and relapse as well as resistance to treatments are associated with glioma stem cells (GSCs). GSCs exhibit a high proliferation rate and self-renewal capacity and the ability to differentiate into diverse cell types, generating a range of distinct cell types within the tumor, leading to cellular heterogeneity. GBM tumors may contain different subsets of GSCs, and some of them may adopt a quiescent state that protects them against chemotherapy and radiotherapy. GSCs enriched in recurrent gliomas acquire more aggressive and therapy-resistant properties, making them more malignant, able to rapidly spread. The impact of SOX transcription factors (TFs) on brain tumors has been extensively studied in the last decade. Almost all SOX genes are expressed in GBM, and their expression levels are associated with patient prognosis and survival. Numerous SOX TFs are involved in the maintenance of the stemness of GSCs or play a role in the initiation of GSC differentiation. The fine-tuning of SOX gene expression levels controls the balance between cell stemness and differentiation. Therefore, innovative therapies targeting SOX TFs are emerging as promising tools for combatting GBM. Combatting GBM has been a demanding and challenging goal for decades. The current therapeutic strategies have not yet provided a cure for GBM and have only resulted in a slight improvement in patient survival. Novel approaches will require the fine adjustment of multimodal therapeutic strategies that simultaneously target numerous hallmarks of cancer cells to win the battle against GBM.
Article
Monoamine oxidase A (MAO A) and heat shock protein 90 (HSP90) inhibitors have been shown to decrease the progression of glioblastoma (GBM) and other cancers. In this study, a series of MAO A/HSP90 dual inhibitors were designed and synthesized in the hope to develop more effective treatment of GBM. Compounds 4-b and 4-c are conjugates of isopropylresorcinol (pharmacophore of HSP90 inhibitor) with the phenyl group of clorgyline (MAO A inhibitor) by a tertiary amide bond substituted with methyl (4-b) or ethyl (4-c) group, respectively. They inhibited MAO A activity, HSP90 binding, and the growth of both TMZ-sensitive and -resistant GBM cells. Western blots showed that they increased HSP70 expression indicating reduced function of HSP90, reduced HER2 and phospho-Akt expression similar to MAO A or HSP90 inhibitor itself. Both compounds decreased IFN-γ induced PD-L1 expression in GL26 cells, suggesting they can act as immune checkpoint inhibitor. Further, they reduced tumor growth in GL26 mouse model. NCI-60 analysis showed they also inhibited the growth of colon cancer, leukemia, non-small cell lung and other cancers. Taken together, this study demonstrates MAO A/HSP90 dual inhibitors 4-b and 4-c reduced the growth of GBM and other cancers, and they have potential to inhibit tumor immune escape.
Article
Full-text available
Optical coherence tomography (OCT) has been recently suggested as a promising method to obtain in vivo and real-time high-resolution images of tissue structure in brain tumor surgery. This review focuses on the basics of OCT imaging, types of OCT images and currently suggested OCT scanner devices and the results of their application in neurosurgery. OCT can assist in achieving intraoperative precision identification of tumor infiltration within surrounding brain parenchyma by using qualitative or quantitative OCT image analysis of scanned tissue. OCT is able to identify tumorous tissue and blood vessels detection during stereotactic biopsy procedures. The combination of OCT with traditional imaging such as MRI, ultrasound and 5-ALA fluorescence has the potential to increase the safety and accuracy of the resection. OCT can improve the extent of resection by offering the direct visualization of tumor with cellular resolution when using microscopic OCT contact probes. The theranostic implementation of OCT as a part of intelligent optical diagnosis and automated lesion localization and ablation could achieve high precision, automation and intelligence in brain tumor surgery. We present this review for the increase of knowledge and formation of critical opinion in the field of OCT implementation in brain tumor surgery.
Article
2098 Background: The prognosis for newly-diagnosed GBM remains poor. Adding temozolomide to radiation therapy improved the median event-free survival (EFS) to 6.9 months and median overall survival to 14.6 months. The five year survival remains < 10%. GBM’s have abundant neo-vascularization and the highest level of vascular endothelial growth factor (VEGF). Bevacizumab is an antibody to VEGF and is the most active agent for recurrent GBM. Hypoxia inducing factor-1 α (HIF-1 α) is an important regulator of VEGF, and topotecan may inhibit HIF-1 α. We performed a phase II trial in newly diagnosed GBM patients, adding bevacizumab and topotecan to standard therapy. Methods: Eighty newly diagnosed GBM patients were enrolled after their craniotomy between December 2009 and December 2010. Patients received standard radiation therapy and temozolomide at 75 mg/m2/d beginning between 2-6 weeks post-craniotomy. Bevacizumab, 10 mg/kg every 14 days was added a minimum of 4 weeks post-op. Two weeks after radiation therap...
Article
OBJECTIVE Gamma knife stereotactic radiosurgery (GK-SRS) is a safe and noninvasive treatment used as adjuvant therapy for patients with glioblastoma multiforme (GBM). Several studies have yielded conflicting results in the effectiveness of radiosurgery in GBM. This study is a retrospective review of our institutional experience with GK-SRS adjuvant therapy in the treatment of GBM. METHODS From October 1998 to January 2003, 51 consecutive patients were treated with GK-SRS as an “upfront” adjuvant therapy after surgery or at the time of tumor progression at Northwestern Memorial Hospital. Survival analysis was performed using the Kaplan-Meier actuarial method. Univariate and multivariate analyses of patient characteristics and treatment variables were performed. RESULTS Treatment with adjuvant GK-SRS yielded a median overall survival of 14.3 months for our cohort. Survival rate of the cohort was 68% at 12 months, 30% at 24 months, and 24% at 36 months. Karnofsky performance score greater than 90 and adjuvant chemotherapy were associated with increased survival on multivariate analysis. Adjuvant GK-SRS performed at tumor progression seems to increase median survival to 16.7 months compared with 10 months when performed after the time of initial tumor resection. Median survival rates by recursive partitioning analysis class breakdown in our cohort are greater than those predicted by other studies. CONCLUSION GK-SRS is a relatively safe and noninvasive procedure that conferred an improvement in overall survival of GBM patients in our retrospective study. Particularly, GK-SRS may improve overall survival when performed at the time of tumor progression.
Article
Autophagy, rather than apoptosis, is induced in gliomas by various treatments, such as radiation and temozolomide (TMZ). The mechanisms of autophagy are gradually being clarified, however, the role of autophagy in cancer therapy remains controversial. Our goal is to elucidate the role of autophagy for application to clinical treatment. We set out to establish methods of monitoring autophagy in in vitro studies and in glioma samples. There are only a few previous reports discussing the detection of autophagy in tissue samples. In vitro studies employing electron microscopy, analysis of LC31/II conversion by western blotting, and GFP-LC3B immunofluorescence assays were undertaken to detect the induction of autophagy. None of these methods alone are sufficient for monitoring autophagy. Therefore, multiple assays to verify an autophagic response were incorporated. The development of a reliable method to monitor autophagy in surgically obtained tissues is vital in elucidating the role of autophagy in the clinical setting. To achieve this goal, immunohistochemistry was utilized in surgical tissue. Macroautophagy was monitored by immunohistochemistry employing anti-LC3B and anti-lysosome associated membrane protein 1 (LAMP1) antibodies. Likewise, chaperone-mediated autophagy was monitored using anti-LAMP2A antibodies. An increase of autophagy after TMZ treatment was noted. LC3B expression by western blotting in frozen tissue samples showed increases in both LC3-I and LC3-II expression after TMZ treatment. By employing immunohistochemical approaches mentioned, it is possible to monitor autophagy in gliomas, opening the door for monitoring therapeutic effects and/or resistance to treatments in gliomas in the clinical setting.
Article
2006 Background: Radiotherapy with concomitant and adjuvant TMZ (TMZ/RT→TMZ) is the standard of care for newly diagnosed GBM. MGMT methylation status may be an important determinant of treatment response. Compared with the standard adjuvant TMZ, dd TMZ results in prolonged depletion of MGMT in blood mononuclear cells and possibly in tumor. This trial determined if intensified TMZ improves survival (OS) or progression free survival (PFS). Methods: This phase III trial was conducted by the RTOG, EORTC and NCCTG. Neurologically stable patients with adequate tissue for prospective MGMT analysis were randomized to Arm 1: standard TMZ (150-200 mg/m2 x 5 d) or Arm 2: dd TMZ (75-100 mg/m2 x 21 d) q 4 wks for 6-12 cycles. Symptom, QOL and neurocognitive testing was performed in a subset of patients. The primary endpoint was OS. Secondary analyses evaluated impact of MGMT status. Eligibility criteria included age > 18 yrs, KPS ≥ 60, and tissue block with > 1cm2 tumor. Results: A total of 833 patients were randomize...
Article
BACKGROUND: In 2004, a randomised phase III trial by the European Organisation for Research and Treatment of Cancer (EORTC) and National Cancer Institute of Canada Clinical Trials Group (NCIC) reported improved median and 2-year survival for patients with glioblastoma treated with concomitant and adjuvant temozolomide and radiotherapy. We report the final results with a median follow-up of more than 5 years. METHODS: Adult patients with newly diagnosed glioblastoma were randomly assigned to receive either standard radiotherapy or identical radiotherapy with concomitant temozolomide followed by up to six cycles of adjuvant temozolomide. The methylation status of the methyl-guanine methyl transferase gene, MGMT, was determined retrospectively from the tumour tissue of 206 patients. The primary endpoint was overall survival. Analyses were by intention to treat. This trial is registered with Clinicaltrials.gov, number NCT00006353. FINDINGS: Between Aug 17, 2000, and March 22, 2002, 573 patients were assigned to treatment. 278 (97%) of 286 patients in the radiotherapy alone group and 254 (89%) of 287 in the combined-treatment group died during 5 years of follow-up. Overall survival was 27.2% (95% CI 22.2-32.5) at 2 years, 16.0% (12.0-20.6) at 3 years, 12.1% (8.5-16.4) at 4 years, and 9.8% (6.4-14.0) at 5 years with temozolomide, versus 10.9% (7.6-14.8), 4.4% (2.4-7.2), 3.0% (1.4-5.7), and 1.9% (0.6-4.4) with radiotherapy alone (hazard ratio 0.6, 95% CI 0.5-0.7; p<0.0001). A benefit of combined therapy was recorded in all clinical prognostic subgroups, including patients aged 60-70 years. Methylation of the MGMT promoter was the strongest predictor for outcome and benefit from temozolomide chemotherapy. INTERPRETATION: Benefits of adjuvant temozolomide with radiotherapy lasted throughout 5 years of follow-up. A few patients in favourable prognostic categories survive longer than 5 years. MGMT methylation status identifies patients most likely to benefit from the addition of temozolomide. FUNDING: EORTC, NCIC, Nélia and Amadeo Barletta Foundation, Schering-Plough.
Article
Background and Purpose: Recurrent malignant gliomas have a very poor prognosis. This trial aimed to evaluate the benefits of reirradiation in case of recurrent glioblastoma multiforme (GBM) using hypofractionated stereotactic radiotherapy (hFSRT) after primary high-dose percutaneous irradiation. Patients and Methods: Between 1998 and 2008, 53 patients with recurrent GBM were treated by hFSRT based on CT and MR imaging. At the time of recurrence, a median total dose of 30 Gy (20–60 Gy) was delivered in median fractions of 3 Gy/day (2–5Gy). Results: The reirradiation was well tolerated (no acute or late toxicity > grade 2), despite the relatively large median tumor volume (35.01 ml). Karnofsky Performance Score was the strongest predictor for survival after reirradiation (p = 0.0159). Tumor volume (p = 0.4690), patient age (p = 0.4301), second operation (p = 0.6930), and chemotherapy (p = 0.1466) at the time of reirradiation did not affect survival. After hFSRT, the median survival was 9 months, and the 1-year progression-free survival (PFS) amounted to 22%.The median overall survival from initial diagnosis was 27 months. 1-year survival from first diagnosis was 83%, 2-year survival 45%. The median time to progression from the end of initial irradiation to recurrence was 12 months. 1-year PFS before reirradiation was 40%. Conclusion: hFSRT as a secondary treatment of recurrent GBM is a feasible and effective treatment option. Only minor side effects were observed with prolonged life expectancy of 9 months.