ArticlePDF AvailableLiterature Review

Therapeutic interactions of autophagy with radiation and temozolomide in glioblastoma: Evidence and issues to resolve

Authors:

Abstract and Figures

Glioblastoma is a unique model of non-metastasising disease that kills the vast majority of patients through local growth, despite surgery and local irradiation. Glioblastoma cells are resistant to apoptotic stimuli, and their death occurs through autophagy. This review aims to critically present our knowledge regarding the autophagic response of glioblastoma cells to radiation and temozolomide (TMZ) and to delineate eventual research directions to follow, in the quest of improving the curability of this incurable, as yet, disease. Radiation and TMZ interfere with the autophagic machinery, but whether cell response is driven to autophagy flux acceleration or blockage is disputable and may depend on both cell individuality and radiotherapy fractionation or TMZ schedules. Potent agents that block autophagy at an early phase of initiation or at a late phase of autolysosomal fusion are available aside to agents that induce functional autophagy, or even demethylating agents that may unblock the function of autophagy-initiating genes in a subset of tumours. All these create a maze, which if properly investigated can open new insights for the application of novel radio- and chemosensitising policies, exploiting the autophagic pathways that glioblastomas use to escape death.British Journal of Cancer advance online publication, 18 February 2016; doi:10.1038/bjc.2016.19 www.bjcancer.com.
Content may be subject to copyright.
Therapeutic interactions of autophagy with
radiation and temozolomide in glioblastoma:
evidence and issues to resolve
Michael I Koukourakis*
,1
, Achilleas G Mitrakas
1
and Alexandra Giatromanolaki
2
1
Department of Radiotherapy/Oncology, Democritus University of Thrace, PO Box 12, Alexandroupolis 68100, Greece and
2
Department of Pathology, Democritus University of Thrace, Alexandroupolis 68100, Greece
Glioblastoma is a unique model of non-metastasising disease that kills the vast majority of patients through local growth, despite
surgery and local irradiation. Glioblastoma cells are resistant to apoptotic stimuli, and their death occurs through autophagy.
This review aims to critically present our knowledge regarding the autophagic response of glioblastoma cells to radiation and
temozolomide (TMZ) and to delineate eventual research directions to follow, in the quest of improving the curability of this
incurable, as yet, disease. Radiation and TMZ interfere with the autophagic machinery, but whether cell response is driven to
autophagy flux acceleration or blockage is disputable and may depend on both cell individuality and radiotherapy fractionation or
TMZ schedules. Potent agents that block autophagy at an early phase of initiation or at a late phase of autolysosomal fusion are
available aside to agents that induce functional autophagy, or even demethylating agents that may unblock the function of
autophagy-initiating genes in a subset of tumours. All these create a maze, which if properly investigated can open new insights
for the application of novel radio- and chemosensitising policies, exploiting the autophagic pathways that glioblastomas use to
escape death.
Glioblastoma is the most common primary brain tumour in adults,
and certainly one of the most lethal human tumours (Hayat et al,
2007) Its incidence is about 3 new cases per 100 000 population per
year, the median age of diagnosis is 64 years and most of the
patients have passed away 2 years following diagnosis (Ohgaki
et al, 2004). Unlike low-grade gliomas, complete surgical removal
of glioblastoma is impossible because of the infiltrative nature of
the disease and the functional vulnerability of the brain. Post-
operative radiotherapy is the standard adjuvant therapy offered
worldwide. Following surgery alone, the median survival is 6
months and increases to 12 months with the addition of
radiotherapy (Salazar et al, 1979). Because of the extent and the
location of the disease adjacent to critical brain areas or because of
the poor performance status of patients at diagnosis, surgery is
often omitted and radiation is offered as the main treatment
option.
Attempts to improve the efficacy of radiotherapy with the
addition of chemotherapy, mainly nitrosoureas (BCNU, CCNU),
go back to the early 1970s. In 1983, Eyre et al in a randomised trial
failed to show a benefit from the addition of CCNU and
procarbazine to radiotherapy. In the same year, an RTOG study
provided evidence that radiochemotherapy with BCNU or methyl-
CCNU or DTIC was superior to radiotherapy alone (Chang et al,
1983). The benefit, however, provided by nitrosoureas was limited
and questionable as stressed in subsequent randomised trials
(Hatlevoll et al, 1985; Deutsch et al, 1989; Shapiro et al, 1989;
Curran et al, 1992). The only therapeutic innovation that had an
impact in the management of glioblastomas appeared in 2005,
when a randomized study showed that temozolomide improved
the median survival of patients undergoing radiotherapy by 2.6
months (Stupp et al, 2005). In a more recent analysis of the
EORTC-NCIC trial on 573 patients, the 2-year survival was 27.2%
in the temozolomide group vs 10.9% in the radiotherapy-alone
group (Stupp, 2009). Nowadays, postoperative radiotherapy with
daily administration of TMZ is the gold standard for patients with
glioblastomas, although the efficacy of this therapy remains
disappointing and attempts to improve the median survival using
the combination of TMZ with other drugs, such as motexafin
*Correspondence: Professor MI Koukourakis; E-mail: targ@her.forthnet.gr
Received 21 September 2015; revised 21 December 2015; accepted 31 December 2015
&2016 Cancer Research UK. All rights reserved 0007 – 0920/16
MINIREVIEW
Keywords: glioblastoma; autophagy; radiotherapy; temozolomide
British Journal of Cancer (2016), 1–12 | doi: 10.1038/bjc.2016.19
www.bjcancer.com | DOI:10.1038/bjc.2016.19 1
Advance Online Publication: 18 February 2016
gadolinium (Brachman et al, 2015), has failed, with the 5-year
survival remaining o10%.
In the era of targeted therapies, there is an increasing optimism
that the identification of key molecular pathways may lead to
major improvements in the management of this highly lethal
disease. Glioblastomas, being highly angiogenic tumours, were
thought to be susceptible to antiangiogenic therapy. Indeed, in
2009, bevacizumab (BVZ), an anti-VEGF humanised monoclonal
antibody, received accelerated approval by the FDA for the
treatment of recurrent glioblastoma following a randomised phase
II study (Kreisl et al, 2009). In the most recent randomised RTOG
0825 trial, however, the addition of BVZ (10 mg kg
1
every 2
weeks) to standard chemoradiation improved the progression-free
interval but had no effect on overall survival (Gilbert et al, 2013).
Recent studies on newly diagnosed glioblastoma patients showed
that the same results and sub-population of patients with better
response to BVZ treatment could not be predicted by the
molecular data (Poulsen et al, 2014). The benefit from BVZ
therapy seems to be confined to oedema reduction and a cut down
on the usage of corticosteroids (Thomas and Omuro, 2014).
Epidermal growth factor receptor-mediated oncogenic signal-
ling is active in most human carcinomas and has become a target
for therapeutic interventions. Indeed, anti-EGFR1 moAb and
pharmacological tyrosine kinase inhibitors have shown important
clinical activities in the colon, lung and head and neck cancer.
About half of glioblastomas bear EGFR gene amplification and
receptor overexpression (Hatanpaa et al, 2010). Mutation of EGFR
with constitutive activation of the pathway is also quite common.
Although the efficacy of such agents as single therapy is limited
(van den Bent et al, 2009), randomised trials in combination with
radiotherapy are missing.
The unique nature of the glioblastoma cells to be resistant to
apoptotic stimuli, their death occurring rather through autophagy
(Yao et al, 2003; Kanzawa et al, 2004), suggests that autophagy-
interfering agents in combination with radiation and temozolo-
mide may represent a new strategy to test. This hypothesis
becomes much more relevant under the new light shed by studies,
suggesting that autophagy is the main pathway exploited by cancer
stem cells to survive and accelerate their renewal (Jiang et al, 2007;
Gong et al, 2013; Berardi et al, 2015). Glioma stem cells is
considered the most radioresistant cell population and autophagy
targeting may assist to eradicate the disease (Shen et al, 2015;
Yu et al, 2015). This review aims to critically present our
knowledge regarding the autophagic response of glioblastoma cells
to radiation and TMZ and to delineate eventual research directions
to follow in the quest of improving the curability of this incurable,
as yet, disease.
AUTOPHAGY AND AUTOPHAGIC DEATH
Macroautophagy (herein stated as autophagy for simplicity) is a
major cell pathway dedicated to a continuous renovation of cell
constituents through the degradation and recycling of long-lived or
damaged proteins and even of entire organelles, such as ribosomes,
mitochondria, endoplasmic reticulum and Golgi apparatus
(Klionsky and Emr, 2000; Feng et al, 2014). During autophagy
double-membrane vesicles are formed, which sequester portions of
the cytoplasm and fuse with endosomal and lysosomal vesicles
(Yorimitsu and Klionsky, 2005). Beclin 1 and ULK (UNC-51-like
kinases) proteins are important autophagosome initiation proteins,
whereas LC3A, LC3B and LC3C are key structural components of
the autophagosomes, which are extensively used for the experi-
mental study of autophagy (Kabeya et al, 2000; Wong et al, 2013),
whereas the difference in expression levels and of subcellular
distribution and kinetics of these proteins may be an indication of
different roles during autophagy (Giatromanolaki et al, 2014;
Koukourakis et al, 2015).
Autophagy-related protein 8 (Atg8/LC3), an ubiquitin-like
protein, has different homologues encoded by different chromo-
somes in animals, including MAP1LC3A, B and C, as well as the
GABARAP proteins, which are the most important constituents of
the autophagosomal membranes (Shpilka et al, 2011). Most of
them are produced as a pre-LC3 25–30 kDa protein that are
cleaved by the autophagophagin (Atg4) to the soluble form of LC3,
the 18 kDa LC3-I protein. Following an ubiquitination-like
reaction, the exposed C terminus conjugates to the head group
amine of phosphatidylethanolamine (PE) through an amide bond,
becoming the so-called membrane-bound 16 kDa LC3-II form that
forms both the outer and inner autophagosome membranes. The
p62/sequestrosome protein is the main carrier of the waste material
to the LC3-II membranes that engulf the complex within a mature
autophagosome (Katsuragi et al, 2015). This is followed by fusion
with lysosomes and digestion of the autophagolysosomal content,
resulting in the disintegration of intracellular waste and at the same
time providing energy to the cell. During this process, LC3-II
protein can be delipidated by Atg4 proteases and liberated in the
cytoplasm. The different forms of LC3 proteins and the kinetics of
the p62 protein can be detected by western blot or confocal
microscopy methods, providing important tools to study the
autolysosomal flux in normal cells and cancer cells under various
stresses (Klionsky et al, 2012).
Under energy-demanding stress conditions, such as starvation
or hypoxia, autophagy is accelerated (Koritzinsky and Wouters,
2013; Bernard et al, 2015). On the one hand, excessive
intensification of this very process may lead to intolerable
cytoplasmic digestion, leading to cell exhaustion and death. On
the other hand, accumulation of autophagosomes under stress,
which block their fusion with lysosomes (e.g., hydroxychloroquine
or 3-methyadenine), may also lead to autophagic death (Kimura
et al, 2013; Saetre et al, 2015). Autophagy is called type II
programmed cell death pathway, which is a distinct pathway
compared with the apoptotic type I death pathway triggered by
DNA or membrane damage followed by release of caspases from
the mitochondria. Autophagy is, therefore, a balance that weighs
between renovation, energy acquisition, survival and cell death,
according to the external or internal stimuli received.
Cancer cells have an increased autophagic activity as shown in a
series of clinicopathological studies, where LC3 and autophagy-
initiating proteins are overexpressed, a feature linked with
aggressive clinical behaviour (Sivridis et al, 2010; Giatromanolaki
et al, 2014; Koukourakis et al, 2015). In a recent study, we noted
that overexpression of LC3A in the form of stone-like cytoplasmic
structures was linked to poor postradiotherapy prognosis in a
series of patients with glioblastoma (Mehta et al, 2015). Since
several decades we know that an inert apoptotic machinery is a
very common phenomenon in cancer cells, allowing them to
escape death after exposure to cytotoxic chemotherapy and
radiotherapy (Hickman, 1996). Evasion from apoptosis is a well-
established phenomenon in gliomas and glioblastomas (Krakstad
and Chekenya, 2010). Therefore, autophagic death is an alternative
pathway to pursue in an attempt to eradicate glioma cells exposed
to cytotoxic stress (Zois and Koukourakis, 2009).
RADIOTHERAPY AND AUTOPHAGY IN GLIOMAS/
GLIOBLASTOMAS
Several experimental studies have confirmed that glioma and
glioblastoma cells exhibit important resistance to apoptosis after
exposure to ionising radiation. Expression status of apoptosis-
related genes such as PTEN may be involved in the phenomenon
(Lee et al, 2011), and interference with various signaling pathways
BRITISH JOURNAL OF CANCER Autophagy, radiation and temozolomide
2 www.bjcancer.com | DOI:10.1038/bjc.2016.19
such as STAT3 and EGFR may restore the ability for apoptotic
death (Gao et al, 2010).
The study by Yao et al (2003) is probably the first to show that
irradiation of human glioblastoma cells results in enhanced
autophagy, whereas no apoptosis was evident in radiosensitive or
radioresistant cell lines. Ito et al (2005) subsequently confirmed
that ionising radiation induces cell cycle arrest and autophagic
death, but not apoptosis, in glioma cell lines. Moreover, Jo et al
(2014) suggested that autophagy is a pathway to cell death after
glioma irradiation. In 2005, a study from the MD Anderson Cancer
Center showed that DNA-protein kinase (DNA-PK)- (enzyme
involved in the repair of DNA double-strand breaks) deficient
glioma cells suffered massive autophagic death even after low doses
of radiation (Daido et al, 2005). Intact DNA-PK pathway
prevented autophagic death, but cells still exhibited a low apoptotic
tendency. Of interest, induction of autophagy using siRNA against
DNA-PK sensitised glioblastoma cell lines (Zhuang et al, 2011a, b).
An additional study using particle radiation, which mainly kills
through DNA double-strand damage, supports the suggestion that
DNA repair disabling is a potent stimulus not only for apoptotic
death but also for autophagic death in gliomas (Jinno-Oue et al,
2010).
The Akt/m-TOR is recognised as a major pathway regulating
autophagy. Inhibitors of Akt/m-TOR activity, such as rapamycin
analogues, intensify the autophagic process (Martelli et al, 2011).
This pathway is often upregulated in tumours, including glioma
(Akhavan et al, 2010). Akt inhibitors induce autophagic death, but
not apoptotic death, in both radioresistant and radiosensitive U87
glioma cell lines and enhance sensitivity to radiation (Fujiwara
et al, 2007). In the same cell line, Benzina et al (2008) found that
high LET radiation kills cells through autophagy. Mehta et al
(2015) further confirmed that Akt inhibition radiosensitises
primary human glioblastoma stem-like cells. Moreover, silencing
of EGFR, an activator of PI3K/Akt/mTOR pathway, with
simultaneous induction of autophagy led to better response to IR
and suppressed migration in the T98G cell line (Palumbo et al,
2014). These results are further supported by Gini et al (2013), who
showed that the growth of EGFRvIII-activated glioblastoma was
blocked after treatment with CC214-1 and CC214-2, which are
inhibitors of mTORC1 and mTORC2. The study by Chiu et al
(2009) suggested that arsenic trioxide also enhances the activity of
radiation in glioma cell lines by augmenting the autophagic cell
death, which is also supported by Carmignani et al (2014), who
showed that glioblastoma stem cells differentiated into non-
tumourigenic cells as a result of autophagy induction, after
inhibition of PI3K/Akt and stimulation of mitogen-activated
protein kinase pathway using arsenic trioxide and metformin,
respectively. Rapamycin has also been shown to induce differ-
entiation of glioma-initiating cells and increase their radio-
sensitivity by activating autophagy (Zhuang et al, 2011a, b).
Furthermore, research related to NF-kB, a factor that suppresses
autophagy in response to TNFa (Djavaheri-Mergny et al, 2007),
showed that treatment of glioma cells with pitavastatin (inhibitor
of NF-kB) resulted in autophagic death but not apoptotic death,
and empowered the activity of radiation (Tsuboi et al, 2009).
Nuclear factor-kB is a transcription factor negatively regulating
apoptotic death. Early phase blockers of autophagy, such as 3-
methyladenine (3-MA), counteracted the cytotoxic effect of
radiation and of the combination with pitavastatin.
A variety of researches focused on glioma radiosensitisation via
autophagy suppression. Yuan et al (2015) exhibited that suppres-
sion of ATG5 using siRNA or suppression of autophagy using 3-
methyladenine increased the radiosensitisation effect of gliomas
after STAT3 inhibition. Moreover, according to Ye et al (2013), the
resistant clones of glioma stem cells bear high expression levels of
early growth response 1 that induce autophagy. Additionally,
mitochondrial isoenzyme of NADP
þ
-dependent isocitrate
dehydrogenase siRNA-transfected A172 glioma cells were sensi-
tised after inhibition of autophagy (Kim et al, 2013).
There is, therefore, important evidence that in glioma cells
autophagic death is the most common pathway exploited by
radiation compared with apoptosis. Although reactive autophagy
may occur in the context of a cell survival response to radiation,
which may represent a radioresistance pathway, augmenting the
autophagic response through combination with Akt/mTOR
inhibitors would shift the balance to death. DNA repair enzyme
inhibitors seem also to facilitate radiation-induced autophagic
death, and at the same time restoring to a certain extent the
apoptotic pathway. The fact that early phase autophagy inhibitors
also sensitise glioma cells to radiation (Lamonaco et al, 2009) is not
contradictory to the whole concept, as abrogating low-level
autophagy that keeps the balance to the survival side is expected
to increase vulnerability to radiation. Late phase autophagy
inhibitors that block autophagolysosomal formation are also
expected to be radiosensitisers as they abrogate cytoprotective
autophagy and load the cell with waste material, resulting in
autophagy-mediated death.
TEMOZOLOMIDE AND AUTOPHAGY IN GLIOMAS/
GLIOBLASTOMAS
One of the first studies regarding the role of autophagy in
malignant gliomas showed that TMZ at clinically relevant
concentration of 100 mM induces autophagy rather than apoptosis
in malignant glioma cells, as shown by the accumulation of the
LC3 protein on autophagosomal membranes. Lee et al (2015)
suggest that TMZ in combination with chloroquine could inhibit
the growth and apoptosis of glioblastomas, whereas autophagy
suppression leads to the abolishment of the combination effect of
TMZ and chloroquine (Lee et al, 2015). Moreover, blockage of
autophagy at an early step by 3-MA prevented the accumulation of
autophagosomes and suppressed the cytotoxic effect of TMZ.
Zanotto-Filho et al (2015), however, suggested that autophagy has
a protective role in gliomas as TMZ/curcumin treatment in
combination with 3-MA leads to a reduction of cell viability.
Nevertheless, bafilomycin that blocks autophagy at a late step, by
preventing the fusion of mature autophagosomes with lysosomes,
sensitised glioma cells to TMZ by inducing apoptotic rather than
autophagic death.
These studies suggest that autophagy and drug interaction is a
very complex process. Blockage of autophagosome formation
abrogates the autophagic death induced by TMZ. On the contrary,
non-functional accumulation of autophagosomes (induced by
TMZ and blocked by late phase autophagy drugs) releases an
apparent pre-existing obstacle in the apoptotic pathway, driving
cells to apoptotic death. Lefranc and Kiss (2006) suggested that as
glioblastoma cells are resistant to apoptosis, blocking targets that
keep autophagy suppressed, such as mTOR, may enhance the
activity of TMZ-induced autophagic death (Zhivotovsky et al,
1999).
An important link between TMZ-induced autophagic death and
cell metabolism has been subsequently brought forward by
Katayama et al (2007). In experiments in multiple glioma cell
lines, TMZ consistently induced autophagy in parallel with an
increase of ATP production. This ATP surge could not be blocked
by glucose starvation but was blocked by agents blocking early
steps of autophagy (methyladenine and beclin 1 siRNA). As this
inhibition resulted in micronucleation, it was suggested that
autophagy-induced ATP surge counteracts the TMZ-induced
autophagic death. Indeed, administration of pyruvate abrogated
the activity of early phase autophagy inhibitors and enhanced
glioma cell survival. Small molecules, such as dasatinib, which
antagonise the ATP binding pocket of abl, kit or EGFR proteins,
Autophagy, radiation and temozolomide BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2016.19 3
induce autophagic cell death in glioblastoma cells and have
synergistic effect with TMZ (Milano et al, 2009). Moreover, the
association between metabolism and mitochondria with the
efficacy of TMZ is supported by a several recent studies. You
et al (2013) showed that the expression of ATAD3A, a
mitochondrial protein ATPase (ATAD3A), is a prognostic factor
for glioblastomas and a predictor of TMZ or radiation resistance
(You et al, 2013). Another study that supports the hypothesis of
association between the function of mitochondria and TMZ
showed that metformin, which inhibits mitochondrial electron
transport chain complex I, enhances the cell cytotoxicity of TMZ
(Sesen et al, 2015).
It seems, therefore, that although exacerbation of autophagy is a
death pathway exploited by TMZ, the energy released by this very
process counteracts TMZ efficacy. Overactivity of the autophagic
pathway in the context of a cell-cytoprotective mechanism against
TMZ can be at the same time the cause of temozolomide-induced
cell death. Abrogation of the end point of such an autophagic-
protective mechanism, which is energy production, inevitably shifts
the balance to increased autophagic death. In pathological studies,
the majority of glioblastoma cases examined had an upregulated
autophagic pathway at diagnosis (Aoki et al, 2008; Pirtoli et al,
2009), with overexpression of LC3, beclin 1 and ULK proteins
(Giatromanolaki et al, 2014), presumably as an indispensable
pathway for energy acquisition under the stressful metabolic
demands of an accelerated growth. Any stressor, like TMZ, that
further accelerates autophagy may result in autophagic death by
disrupting a delicate balance between energy replenishment and
suicide autophagy. The synergism found between TMZ-induced
autophagy and autophagy-mediated killing effect of oncolytic
adenoviruses (Yokoyama et al, 2008; Ulasov et al, 2009) may also
be explained under this point of view.
An interesting observation from the clinicopathological studies
is that a glioblastoma group with low levels of LC3 and beclin 1 has
a poorer prognosis and may be more resistant to TMZ, although
this hypothesis demands further confirmation in larger studies.
A study by Fu et al (2009) provides important insights into
this phenomenon. Following separation of CD133-positive
and -negative glioblastoma cells from a freshly resected tumour,
positive cells (representing glioblastoma stem cells) had lower
LC3-II and beclin 1 levels compared with negative ones and were
more resistant to TMZ (Fu et al, 2009). Both cells, however, were
resistant to caspase-3 induction following incubation with TMZ.
An important hypothesis that emerges from these studies is that,
although autophagic death is a main pathway of glioma cell death
after exposure to TMZ, glioma stem cells may have an autophagic
machinery refractory to the activity of the drug and, therefore,
more easily survive under exposure to TMZ, leading subsequently
to tumour repopulation and growth.
The above data contrast the study by Lomonaco et al (2009)
where CD133-positive cells seem to upregulate autophagy more
potently than negative cells after exposure to radiation and to the
finding that autophagy inhibition sensitises the stem cell sub-
population to radiation. Similar results have been reported by
Winardi et al (2014), who supports that high level of CD133
population and high rate of autophagy lead to a poor prognosis in
astrocytomas (Winardi et al, 2014). Although further studies are
demanded to elucidate this discrepancy that may be cancer cell line-
dependent, differences in the mechanism of autophagy induction
between radiation and TMZ can also underlie. In addition, it is
stressed that for currently applied experimental techniques, it is
difficult to distinguish between autophagosomal or autolysosomal
non-functional accumulation vs intensified autophagy. Moreover, as
LC3A and LC3B are distinct proteins with different response at
various stimuli, studying the autophagic response with nonspecific
LC3 subtype markers may provide confusing results and mask
specific response patterns that may be important to TMZ activity.
In any case, restoration or protection of the autophagic flux may
be important in sensitising cells to TMZ. Methylation of the
promoter of Beclin 1 gene has been reported to be a common event
in breast cancer (Li et al, 2010). Whether hypomethylating
agents can be of value to restore autophagic responsiveness in
glioma stem cells and sensitivity to TMZ is a sound hypothesis.
5-Aza-20-deoxycytidine, a potent demethylating agent, is an
autophagy promoter (Chen et al, 2011). Although it is unknown
whether demethylating agents can restore autophagy in glioblas-
tomas, we know that they restore the expression of genes involved
in apoptosis (Eramo et al, 2005) like caspase-8, which is
hypermethylated in stem cell-like glioma cells (Capper et al,
2009). However, differentiation-inducing agent inhibitors of
histone deacetylase, such as valproic acid (Park et al, 2011), are
potent inducers of autophagy in glioma cells, enhancing auto-
phagic cell death but not apoptosis (Fu et al, 2010). Whether such
agents can target TMZ-resistant glioma stem cells is another
hypothesis to test. Carbamazepine is also another drug that
increases the autophagic flux and protects normal tissues against
radiation (Kim et al, 2011), presumably due to restoration of the
radiation-induced autophagy blockage (Zois et al, 2011).
RESTORING APOPTOSIS
Another important approach aside to autophagy death pathway
manipulation is to interfere with the molecular pathways aiming to
overcome the relative resistance of glioma cells to apoptotic
death. Both radiotherapy and TMZ are DNA-damaging agents.
Radiotherapy induces both DNA single- and double-strand breaks,
the kinetics and fidelity of repair of which define cell survival and
clonogenic ability (Mirzayans et al, 2013; Morgan and Lawrence,
2015). Whether apoptosis, mitotic catastrophe or senescence
follows defective DNA repair is an outcome defined, at least
partially, by the oncogene and apoptosis-related gene activity in
cancer cells (Vakifahmetoglu et al, 2008; Mirzayans et al, 2013).
Gene therapy or small molecules restoring p53 function increase
the radiation-induced apoptosis in cancer cells, as well as in
glioblastomas (Villalonga-Planells et al, 2011; Pflaum et al, 2014).
Temozolomide is also a DNA-damaging agent. Following its
conversion to 5-aminoimidazole-4-carboxamide, it delivers methyl
groups to DNA, resulting in the formation of O
6
-methylguanine,
which mispairs erroneously with thymine (instead of cytosine)
during DNA replication, resulting in DNA double-strand break
formation (Fukushima et al, 2009). The death pathway that follows
depends on the gene expression profile of the cancer cell, the p53,
PUMA and bcl-2 family of protein expression status being decisive
for the activation of the apoptosis pathway under exposure to
temozlomide, even of glioma stem cells (Gratas et al, 2014; Miao
et al, 2015).
Gene therapy or novel therapeutic agents that restore the
apoptotic machinery in gliomas may prove to be of importance in
reactivating apoptotic death induced by both ionising radiation
and TMZ. Inhibitors of the PDGFR seem to initiate the apoptotic
pathway in apoptosis-resistant glioblastoma cells (Ziegler et al,
2008). Anti-EGFR therapy has been reported to sensitise CD133-
positive glioma cells to radiation, presumably by restoring their
apoptotic ability (Diaz Miqueli et al, 2009). Specific inhibitors of
the checkpoint kinases Chk1 and Chk2 reverse the radioresistane
of these cells (Bao et al, 2006). Drugs targeting the mitochondrial
pore, such as lonidamine, have been shown to induce apoptotic
death in TMZ-resistant glioma cells (Lena et al, 2009). Chang et al
(2009) found that Sirtuin 1 gene is exclusively expressed in CD133-
positive radioresistant stem cells and that silencing this gene
improves curability in experimental models. Agonist antibodies
of the TRAIL death receptor 5 that induce apoptosis are
BRITISH JOURNAL OF CANCER Autophagy, radiation and temozolomide
4 www.bjcancer.com | DOI:10.1038/bjc.2016.19
also shown to improve the efficacy of radiotherapy and TMZ
(Fiveash et al, 2008). Gene therapy approaches, such as introduc-
tion of the caspase-8 gene or IL-24 gene, may be also useful in
restoring apoptosis of glioma cells (Tsurushima et al, 2008; Yacoub
et al, 2008).
WHICH AUTOPHAGIC DEATH?
The above data suggest that glioma and glioblastoma cells exhibit
an autophagic response after exposure to ionising radiation and
TMZ. This assumption is based on the increased accumulation of
LC3-positive autophagosomes. Such a finding, however, does not
necessarily mean an intensified ‘functional autophagy’. In fact,
disruption of the lysosomal and autophagosomal fusion may also
result in LC3-positive autophagosomal accumulation, without
intensification of the production of new autophagosomes. This is
an important point we need to clarify regarding the actual effect of
ionising radiation on the autophagic machinery. Autophagic flux
studies in cancer cells are demanded to elucidate the phenomenon,
and such studies are absent in the literature. Our studies in normal
fibroblasts and endothelial cells suggest that radiation results in an
early blockage of the autophagic flux within the first days of
irradiation (Kalamida et al, 2015). The increase of LC3-II
membrane-bound form in the soluble fraction of cells instead of
the pellet fraction and the sharp increase of p62 protein (that
normally disintegrates in the autophagolysosomal environment
once incorporated) strongly suggest that radiation induces a non-
functional accumulation of autophagosomes in cells, inducing
autophagic death in a similar way that late phase autophagy
blockers (such as chloroquine and bafilomycin) do. In fact,
restoration of autopagic flux protects normal cells from radiation
death.
Whether this phenomenon also applies in cancer cells or
whether these sustain a resistant attitude against radiation-induced
autophagy flux blockage demands thorough investigation. Pre-
liminary experiments show that the response of cancer cells seems
to be cell line- and radiation dose-specific (unpublished data).
A biphasic relation linking radiation dose levels and the autophagic
survival/death balance in cancer cells cannot be excluded. Lower
doses, especially in radioresistant cells, could trigger a functional
autophagic response with a rather cytoprotective role providing
energy to the cells. In contrast, higher doses may disrupt the
autophagic process by downregulating autophagy gene expression
and/or by blocking autophagosome/lysosomal interaction shifting
the balance to an autophagic death because of an accumulation of
waste. This, however, is nothing more than a hypothesis that
should be thoroughly examined, as different fractionation of
radiotherapy may have a different effect on autophagy and may
synergise better with different autophagy-interfering agents.
Nevertheless, in a recent study in prostate cancer we provided a
strong evidence that such a hypothesis is valid, as a radioresistant
cell line intensified its autophagic flux after low-dose radiation,
whereas this was blocked in a radiosensitive one using the same
radiation dose (Koukourakis et al, 2015). It may be, therefore, that
radioresistant tumours continue or accelerate their autophagic flux
when exposed to low dose per fraction (i.e. within the range of
standard 2 Gy per day fractionation), whereas larger fractions
(hypofractionation) may block the autophagic flux and at least
partially overcome radioresistance. Silencing of the LC3A gene
results in important radiosensitisation, suggesting that autophagy
is eventually a pathway of survival following irradiation of
radioresistant cells and tumours (Koukourakis et al, 2015).
Nowadays, it is more than clear that autophagy is overactive in a
large fraction of tumours, including gliomas (Capper et al, 2009),
and cell death features representative of an autophagic exhaustion
of the cytoplasmic material are commonly evident in the form of
stone-like cytoplasmic structures. Indeed, we identified the
formation of the so-called LC3A-positive ‘stone-like structures’ in
various cancer tissues, showing a complete elimination of the
cytoplasm of cells and its substitution by one or more dense
structures of amorphous undigested LC3-positive material (Sivridis
et al, 2010). This feature, also evident in human glioblastomas, was
inducible under acidic conditions and glucose antagonism,
showing the existence of this type of autophagic cell death in
glioblastomas (Capper et al, 2009). This autophagic death path
presumably demands functional and overactivated autophagy to
uncontrollably consume the cytoplasmic content, shifting the
balance from autophagic cytoprotection to autophagic death. Thus,
as cancer cells use functional upregulated autophagy to survive the
hypoxic and overall unfavorable tumour environment, further
exogenous induction of functional autophagy, for example,
through Akt/mTOR inhibitors, may push the balance towards
cytoplasmic exhaustion and autophagic death.
It can be, therefore, suggested that autophagic death is not a
single phenomenon but is rather characterised by at least two
distinct paths: (i) the excessive autophagic activity resulting in
stone-like structure formation (functional autophagic death) by
exhaustion of the lysosomal potential and; (ii) the intolerable
accumulation of waste non-functional autophagosomes (non-
functional autophagic death) by abrogation of lysosomal fusion.
Whether this latter path represents a link of autophagy with
apoptosis, necrosis or mitotic catastrophe induction or it represents
an entirely distinct death pathway is unknown.
THE PARADOXICAL SENSITISATION BY BOTH INDUCERS
AND BLOCKERS
Nevertheless, both ‘functional’ and ‘non-functional’ autophagic
death pathways triggered by radiation and TMZ can be boosted by
early or late phase autophagy blockers and by autophagy inducers.
This paradox can be explained as follows:
(1) Early phase autophagy blockers prevent the formation of
autophagosomes, blocking therefore an important source of
energy and the cytoprotective effect of a controllable functional
autophagy. This, on the one hand, leads per se to the death of a
varying fraction of tumour cells, the survival of which
depended on functional autophagy. On the other hand, by
blocking autophagosome formation tumour cells enter a state
of energy deficit and excessive loading with damaged proteins
and organelles that radiation or TMZ produces, facilitating the
apoptotic death effect of these agents. Thus, early autophagy
blockers may activate a link with apoptosis-like pathways. For
such therapeutic approaches, apoptosis restoration policies
may secure a superadditive effect.
(2) Late phase autophagy blockers, for example, those that allow
the formation of autophagosomes but block their fusion with
lysosomes, repress by one hand the acquisition of energy
through autophagy and on the other hand load the cell with
waste non-degradable autophagic vesicles. Thus, similarly to
early phase blockers, they kill a varying percentage of tumour
cells through energy deprivation and waste overloading. If a
radiotherapy schedule and/or TMZ also induces non-func-
tional autophagosome formation, the synergistic effect with the
energy austerity effect or with the autophagosome accumula-
tion is antagonistic or at least additive. However, if a
radiotherapy schedule is given in those small fractions to
induce functional autophagy, the intensified accumulation of
autophagosomes using late blockers would result in super-
additive effect. In fact, a combination of an early phase
autophagy inducer with a late phase blocker seem an appealing
therapeutic proposal.
Autophagy, radiation and temozolomide BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2016.19 5
(3) Autophagy inducers could not only target tumour cells with
increased autophagy but also tumour cells with reduced
autophagy. In tumour cells with active autophagy, augmentation
of autophagy may shift the balance to functional autophagic
death through lysosomal exhaustion and cytoplasmic degrada-
tion (stone-like death). If radiation and TMZ is scheduled to
induce functional autophagy, autophagy inducers administered
concurrently with radiation will accelerate functional autophagic
death. If radiation and TMZ are scheduled to induce non-
functional autophagosome accumulation, administration of
autophagy inducers immediately before radiochemotherapy
may enhance the non-functional autophagic death.
CLINICAL EXPERIENCE
Clinical experience with autophagy-manipulating agents in the
treatment of glioblastoma is limited. Chloroquine (Kimura et al,
2013), presumably because it is already availabile in the clinical
practice as an antimalaria agent (Hall, 1976), is the only autophagy
inhibitor studied for the treatment of glioblastoma patients. In a
phase II clinical trial, the administration of hydroxychloroquine to
patients with glioblastoma undergoing radiotherapy with temozo-
lomide confirmed an increase of autophagic vacuoles and of the
LC3-II form in the peripheral blood cells, supportive of an
antiautophagic activity, but the haematological toxicity of the regimen
was unacceptable and the benefit in terms of survival was not evident
(Rosenfeld et al, 2014). A small pilot study on the combination of
chloroquine with radiation for the treatment of recurrent glioblas-
toma confirmed the feasibility of the regimen and the authors claimed
encouraging treatment outcomes (Bilger et al, 2014). The recent
observation that the acidic extracellular pH neutralises the autophagy-
inhibiting activity of chloroquine stresses the importance of the
development of potent autophagy inhibitors, the activity of which is
independent or even better take advantage of the intratumoral
hypoxic and acidic conditions (Pellegrini et al, 2014).
Mammalian target of rapamycin inhibitors have multiple
biological activities, including the induction of autophagy
(Huang and Fingar, 2014). Such agents have been introduced in
the clinical practice for the treatment of renal carcinoma (Amato,
2011), and several clinical trials have investigated the activity
mTOR inhibitors in patients with glioblastoma. Temsirolimus, for
example, showed clinical efficacy in one-third of patients treated
for recurrent glioblastoma (Galanis et al, 2005). Sarkaria et al
(2011) combined everolimus in combination with radiotherapy
and TMZ in 18 patients, showing a metabolic effect of everolimus
in most patients, as detected with fluorodeoxyglucose PET scan.
More recently, the authors reported a phase II study on 100
patients, which, however, did not detect any beneficial effect in
terms of survival (Ma et al, 2015). An interesting phase II study by
Hainsworth et al (2012) administered BVZ with everolimus after
radiochemotherapy for 68 glioblastoma patients (Hainsworth et al,
2012). The progression-free survival interval compared favorably
with the authors’ previous experience.
IMPORTANT ISSUES TO RESOLVE
It is evident that although we know that radiation and TMZ kill
cells through autophagy, it is unclear which of the functional or
non-functional autophagic death pathway is followed. It is very
important to investigate the role of radiotherapy fractionation on
the autophagic pathways that this activates. Several tumours, such
as gliomas, melanomas or even prostate cancer, exhibit a higher
sensitivity to large radiotherapy fractions. It may be that small
radiotherapy fractions (around 2 Gy) can simply stimulate
functional autophagy in such cells, triggering therefore a
cytoprotective pathway during the fractionated course of radio-
therapy. Larger fractions can block autophagic flux leading to non-
functional accumulation of autophagosomes, so that in such
radiation scheme self-sensitises cells to its effect.
If such a differential effect on autophagy exists, then the choice
and sequence of autophagy-interfering agent for combined
administration should be made according to the fractionation of
radiotherapy we wish to apply. The same principle can be also
applicable for TMZ as the short schedule of 200–250 mg m
2
5
days every month or the prolonged schedule of 70 mg m
2
per day
continuously used in the clinical practice may have a different
effect on the autophagic flux of gliomas and may demand different
autophagic interference to achieve radio/chemosensitisation.
It seems that the interfering autophagy may indeed help us
make a difference in the treatment of gliomas and glioblastomas. A
hypothetical scheme for intervention targeting autophagy aiming
to radiosensitisation is shown in Figure 1. Inducers of autophagy,
like mTOR inhibitors available in the clinical practice (Houghton,
2010), can lead to a functional autophagic death when combined
with standard radiotherapy and a schedule of continuous low daily
dose of TMZ. Pretreatment of tumours with autophagy inducers,
followed by hypofractionated radiotherapy and or high daily dose
schedules of TMZ, in combination with late phase autophagy
blockers, could enhance antineoplastic efficacy by leading to non-
functional autophagic death. Whether early phase autophagy
blockers could be of value in the combination of agents that restore
apoptotic death pathways with radiotherapy or TMZ is also a
hypothesis to study. Important issues remain, however, unresolved.
One key question that has to be answered is whether radiotherapy
and TMZ trigger a functional autophagic response or they just
deregulate autophagy resulting in non-degradable autophagosome
accumulation. This demands studies of monitoring the autophagic
flux and lysosomal kinetics in cell lines after escalated doses of
radiotherapy or TMZ, as dose schedule may be a principal factor
defining the type of autophagic response.
Another important issue to resolve is the characterisation of the
autophagic activity and response tendency of the tumour itself. Not
all gliomas or glioblastomas are the same as, indeed, some of them
are more radiosensitive than others. Assessment of the expression
status of autophagy-blocking pathways such as Akt/mTOR may
help to identify tumours of defective autophagy, the radio-
sensitivity of which may increase by specific Akt/mTOR inhibitors.
As beclin 1 protein is downregulated in a large percentage of
gliomas, detection of hypermethylated Beclin 1 gene or other
autophagy-initiating proteins may reveal targets for demethylating
agent administration so that subsequent interference with
autophagy inducers may become beneficial. Extensive expression
of LC3A-positive stone-like structures may characterise tumours
with functional autophagy that may benefit from concurrent
radiotherapy with either early autophagy and DNA-repair blockers
or autophagy induction therapy with mTOR inhibitors (depending
upon radiotherapy fractionation).
Assessment of the content of CD133-positive compartment can
be easily performed in immunohistochemistry of biopsies.
Pretreatment of these patients with differentiation-inducing agents
such as histone deacetylase inhibitors may prove a useful
preradiotherapy policy. Silencing of stem-cell-specific genes, such
as Sirtuin 1, or apoptosis restoration treatments, such as caspase or
IL-24 adenoviruses, may also be of value in these cases.
CONCLUSION
Malignant glioma is probably a unique model of non-metastasising
disease that kills the majority of patients through local growth.
BRITISH JOURNAL OF CANCER Autophagy, radiation and temozolomide
6 www.bjcancer.com | DOI:10.1038/bjc.2016.19
It consists, therefore, of one of the biggest challenges in radiation
therapy, especially nowadays when novel radiotherapy techniques
allow the delivery of high dose per fraction to the tumour at the
same time reducing the dose to the surrounding normal brain
tissue. Whether hypofractionation that would block the autophagy
flux, even in radioresistant cell lines, can be effectively combined
with late autophagy blockers to exploit a non-functional autopha-
gic death is a question that deserves investigation. Temozolomide
is an important drug when combined with radiotherapy, or even
for relapsed tumours after radiotherapy, and just like radiotherapy
kills glioma cells through autophagic death. The combination of
mTOR inhibitors (intensifiers or autophagy) with low daily dose of
TMZ during standard radiotherapy or after failure of radiotherapy,
aiming to trigger functional autophagic death, may also be a
promising approach. Modulation of radiation or of TMZ
autophagic response appears one of the most promising
approaches to prolong survival and to better understand the
glioblastoma therapy riddle, but important insights demanded to
create a reliable concept are still missing.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
REFERENCES
Akhavan D, Cloughesy TF, Mischel PS (2010) mTOR signaling in
glioblastoma: lessons learned from bench to bedside. Neuro Oncol 12:
882–889.
Amato R (2011) Everolimus for the treatment of advanced renal cell
carcinoma. Expert Opin Pharmacother 12: 1143–1155.
Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T,
Hollingsworth EF, Kobayashi R, Hess K, Shinojima N, Shingu T,
Tamada Y, Zhang L, Conrad C, Bogler O, Mills G, Sawaya R, Kondo S
(2008) Monitoring autophagy in glioblastoma with antibody against
isoform B of human microtubule-associated protein 1 light chain 3.
Autophagy 4: 467–475.
Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW,
Bigner DD, Rich JN (2006) Glioma stem cells promote radioresistance
by preferential activation of the DNA damage response. Nature 7:
756–760.
Benzina S, Altmeyer A, Malek F, Dufour P, Denis JM, Gueulette J, Bischoff P
(2008) High-LET radiation combined with oxaliplatin induce autophagy
in U-87 glioblastoma cells. Cancer Lett. 8: 63–70.
Berardi DE, Flumian C, Rodriguez CE, Dı
´az Bessone MI, Cirigliano SM,
Bal de Kier Joffe
´ED, Fiszman GL, Urtreger AJ, Todaro LB (2015)
PKCdinhibition impairs mammary cancer proliferative capacity but
selects cancer stem cells, involving autophagy. J Cell Biochem 9999:
1–11.
Bernard A, Jin M, Xu Z, Klionsky DJ (2015) A large-scale analysis of
autophagy-related gene expression identifies new regulators of autophagy.
Autophagy 11: 2114–2122.
Bilger A, Bittner MI, Grosu AL, Wiedenmann N, Meyer PT, Firat E,
Niedermann G, Weber WA, Milanovic
´D (2014) FET-PET-based
reirradiation and chloroquine in patients with recurrent glioblastoma:
first tolerability and feasibility results. Strahlenther Onkol 190:
957–961.
Brachman DG, Pugh SL, Ashby LS, Thomas TA, Dunbar EM, Narayan S,
Robins HI, Bovi JA, Rockhill JK, Won M, Curran WP (2015) Phase 1/2
trials of temozolomide, motexafin gadolinium, and 60-Gy fractionated
radiation for newly diagnosed supratentorial glioblastoma multiforme:
final results of RTOG 0513. Int J Radiat Oncol Biol Phys. 1: 961–967.
Capper D, Gaiser T, Hartmann C, Habel A, Mueller W, Herold-Mende C, von
Deimling A, Siegelin MD (2009) Stem-cell-like glioma cells are resistant to
TRAIL/Apo2L and exhibit down-regulation of caspase-8 by promoter
methylation. Acta Neuropathol 117: 445–456.
Carmignani M, Volpe AR, Aldea M, Soritau O, Irimie A, Florian IS,
Tomuleasa C, Baritchii A, Petrushev B, Crisan G, Valle G (2014)
Glioblastoma stem cells: a new target for metformin and arsenic trioxide.
J Biol Regul Homeost Agents 28: 1–15.
Chang CH, Horton J, Schoenfeld D (1983) Comparison of postoperative
radiotherapy and combined postoperative radiotherapy and chemotherapy
in the multidisciplinary management of malignant gliomas. A joint
Radiation Therapy Oncology Group and Eastern Cooperative Oncology
Group study. Cancer 52: 997–1007.
PIK3/Akt/mTOR inhibitors
autophagy inducers
Small fractionation of RT
Low-dose intensity temozolomide
(inducers of functional autophagy)
Functional
autophagic
death
Pretreatment with
PIK3/Akt/mTOR
inhibitors to further
intensify autophagy
Large fractionation RT
High-dose intensity
temozolomide
(accumulation of
autophagosomes)
Late-phase
autophagy blockers
Non-functional
autophagic
death
Apoptopic death
RT+temozolomide
(DNA damage effect)
Apoptosis restoration
agents
Early-phase
autophagy blockers
Demethylating agents
Gliomas with increased or
functional autophagic activity
Gliomas with repressed
autophagic activity
Figure 1. A hypothetical scheme targeting autophagy for glioblastoma sensitisation during radio-chemotherapy with TMZ.
Autophagy, radiation and temozolomide BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2016.19 7
Chang CJ, Hsu CC, Yung MC, Chen KY, Tzao C, Wu WF, Chou HY,
Lee YY, Lu KH, Chiou SH, Ma HI (2009) Enhanced radiosensitivity
and radiation-induced apoptosis in glioma CD133-positive cells by
knockdown of SirT1 expression. Biochem Biophys Res Commun 6:
236–242.
Chen MY, Liao WS, Lu Z, Bornmann WG, Hennessey V, Washington MN,
Rosner GL, Yu Y, Ahmed AA, Bast RC Jr (2011) Decitabine and
suberoylanilide hydroxamic acid (SAHA) inhibit growth of ovarian cancer
cell lines and xenografts while inducing expression of imprinted tumour
suppressor genes, apoptosis, G2/M arrest, and autophagy. Cancer 117:
4424–4438.
Chiu HW, Ho SY, Guo HR, Wang YJ (2009) Combination treatment with
arsenic trioxide and irradiation enhances autophagic effects in U118-MG
cells through increased mitotic arrest and regulation of PI3K/Akt and
ERK1/2 signaling pathways. Autophagy 5: 472–483.
Curran Jr WJ, Scott CB, Nelson JS (1992) A randomized trial of accelerated
hyperfractionated radiation therapy and bis-chloroethyl nitrosourea for
malignant glioma. A preliminary report of Radiation Therapy Oncology
Group 83-02. Cancer 70: 2909–2917.
Daido S, Yamamoto A, Fujiwara K, Sawaya R, Kondo S, Kondo Y (2005)
Inhibition of the DNA-dependent protein kinase catalytic subunit
radiosensitizes malignant glioma cells by inducing autophagy. Cancer Res
65: 4368–4375.
Deutsch M, Green SB, Strike TA (1989) Results of a randomized trial
comparing BCNU plus radiotherapy, streptozotocin plus radio-
therapy, BCNU plus hyperfractionated radiotherapy, and BCNU
following misonidazole plus radiotherapy in the postoperative
treatment of malignant glioma. IntJRadiatOncolBiolPhys16:
1389–1396.
Diaz Miqueli A, Rolff J, Lemm M, Fichtner I, Perez R, Montero E (2009)
Radiosensitisation of U87MG brain tumours by anti-epidermal
growth factor receptor monoclonal antibodies. Br J Cancer 100:
950–958.
Djavaheri-Mergny M, Amelotti M, Mathieu J, Besanc¸on F, Bauvy C,
Codogno P (2007) Regulation of autophagy by NFkappaB transcription
factor and reactives oxygen species. Autophagy 3: 390–392.
Eramo A, Pallini R, Lotti F, Sette G, Patti M, Bartucci M, Ricci-Vitiani L,
Signore M, Stassi G, Larocca LM, Crino
`L, Peschle C, De Maria R (2005)
Inhibition of DNA methylation sensitizes glioblastoma for tumour
necrosis factor-related apoptosis-inducing ligand-mediated destruction.
Cancer Res 65: 11469–11477.
Eyre HJ, Quagliana JM, Eltringham JR (1983) Randomized comparisons of
radiotherapy and CCNU versus radiotherapy, CCNU plus procarbazine
for the treatment of malignant gliomas following surgery. A Southwest
Oncology Group Report. J Neurooncol 1: 171–177.
Feng Y, He D, Yao Z, Klionsky DJ (2014) The machinery of macroautophagy.
Cell Res 24: 24–41.
Fiveash JB, Gillespie GY, Oliver PG, Zhou T, Belenky ML, Buchsbaum DJ
(2008) Enhancement of glioma radiotherapy and chemotherapy response
with targeted antibody therapy against death receptor 5. Int J Radiat Oncol
Biol Phys. 71: 507–516.
Fu J, Liu ZG, Liu XM, Chen FR, Shi HL, Pangjesse CS, Ng HK, Chen ZP
(2009) Glioblastoma stem cells resistant to temozolomide-induced
autophagy. Chin Med J (Engl) 122: 1255–1259.
Fu J, Shao CJ, Chen FR, Ng HK, Chen ZP (2010) Autophagy induced by
valproic acid is associated with oxidative stress in glioma cell lines.
Neuro-Oncology 12: 328–340.
Fujiwara K, Iwado E, Mills GB, Sawaya R, Kondo S, Kondo Y (2007) Akt
inhibitor shows anticancer and radiosensitizing effects in malignant
glioma cells by inducing autophagy. Int J Oncol 31: 753–760.
Fukushima T, Takeshima H, Kataoka H (2009) Anti-glioma therapy with
temozolomide and status of the DNA-repair gene MGMT. Anticancer Res
29: 4845–4854.
Galanis E, Buckner JC, Maurer MJ, Kreisberg JI, Ballman K, Boni J,
Peralba JM, Jenkins RB, Dakhil SR, Morton RF, Jaeckle KA,
Scheithauer BW, Dancey J, Hidalgo M, Walsh DJ. North Central Cancer
Treatment Group (2005) Phase II trial of temsirolimus (CCI-779) in
recurrent glioblastoma multiforme: a North Central Cancer Treatment
Group Study. J Clin Oncol 23: 5294–5304.
Gao L, Li F, Dong B, Zhang J, Rao Y, Cong Y, Mao B, Chen X (2010)
Inhibition of STAT3 and ErbB2 suppresses tumour growth, enhances
radiosensitivity, and induces mitochondria-dependent apoptosis in glioma
cells. Int J Radiat Oncol Biol Phys 77: 1223–1231.
Giatromanolaki A, Sivridis E, Mitrakas A, Kalamida D, Zois CE, Haider S,
Piperidou C, Pappa A, Gatter KC, Harris AL, Koukourakis MI (2014)
Autophagy and lysosomal related protein expression patterns in human
glioblastoma. Cancer Biol Ther 15: 1468–1478.
Gilbert MR, Dignam J, Won M, Blumenthal DT, Vogelbaum MA, Aldape KD,
Colman H, Chakravarti A, Jeraj R, Armstrong TS, Wefel JS, Brown PD,
Jaeckle KA, Schiff D, Atkins JN, Brachman D, Werner-Wasik M,
Komaki R, Sulman EP, Mehta MP (2013) A randomized trial of
bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370:
699–708.
Gini B, Zanca C, Guo D, Matsutani T, Masui K, Ikegami S, Yang H,
Nathanson D, Villa GR, Shackelford D, Zhu S, Tanaka K, Babic I,
Akhavan D, Lin K, Assuncao A, Gu Y, Bonetti B, Mortensen DS, Xu S,
Raymon HK, Cavenee WK, Furnari FB, James CD, Kroemer G, Heath JR,
Hege K, Chopra R, Cloughesy TF, Mischel PS (2013) The mTOR kinase
inhibitors, CC214-1 and CC214-2, preferentially block the growth of
EGFRvIII-activated glioblastomas. Clin Cancer Res 19: 5722–5732.
Gong C, Bauvy C, Tonelli G, Yue W, Delome
´nie C, Nicolas V, Zhu Y,
Domergue V, Marin-Esteban V, Tharinger H, Delbos L, Gary-Gouy H,
Morel AP, Ghavami S, Song E, Codogno P, Mehrpour M (2013)
Beclin 1 and autophagy are required for the tumorigenicity of breast
cancer stem-like/progenitor cells. Oncogene 32: 2261–2272.
Gratas C, Se
´ry Q, Rabe
´M, Oliver L, Vallette FM (2014) Bak and Mcl-1 are
essential for Temozolomide induced cell death in human glioma.
Oncotarget 5: 2428–2435.
Hainsworth JD, Shih KC, Shepard GC, Tillinghast GW, Brinker BT, Spigel DR
(2012) Phase II study of concurrent radiation therapy, temozolomide, and
bevacizumab followed by bevacizumab/everolimus as first-line treatment
for patients with glioblastoma. Clin Adv Hematol Oncol 10: 240–246.
Hall AP (1976) The treatment of malaria. BMJ 1: 323–328.
Hatanpaa KJ, Burma S, Zhao D, Habib AA (2010) Epidermal growth factor
receptor in glioma: signal transduction, neuropathology, imaging, and
radioresistance. Neoplasia 12: 675–684.
Hatlevoll R, Lindegaard KF, Hagen S (1985) Combined modality treatment of
operated astrocytomas grade 3 and 4. A prospective and randomized study
of misonidazole and radiotherapy with two different radiation schedules
and subsequent CCNU chemotherapy. Stage II of a prospective
multicenter trial of the Scandinavian Glioblastoma Study Group. Cancer
56: 41–47.
Hayat MJ, Howlader N, Reichman ME, Edwards BK (2007) Cancer statistics,
trends, and multiple primary cancer analyses from the Surveillance,
Epidemiology, and End Results (SEER) Program. Oncologist 12: 20–37.
Hickman JA (1996) Apoptosis and chemotherapy resistance. Eur J Cancer
32A: 921–926.
Houghton PJ (2010) Everolimus. Clin Cancer Res 16(5): 1368–1372.
Huang K, Fingar DC (2014) Growing knowledge of the mTOR signaling
network. Semin Cell Dev Biol 36: 79–90.
Ito H, Daido S, Kanzawa T, Kondo S, Kondo Y (2005) Radiation-induced
autophagy is associated with LC3 and its inhibition sensitizes malignant
glioma cells. Int J Oncol 26: 1401–1410.
Jiang H, Gomez-Manzano C, Aoki H, Alonso MM, Kondo S, McCormick F,
Xu J, Kondo Y, Bekele BN, Colman H, Lang FF, Fueyo J (2007)
Examination of the therapeutic potential of Delta-24-RGD in brain
tumor stem cells: role of autophagic cell death. J Natl Cancer Inst 99:
1410–1414.
Jinno-Oue A, Shimizu N, Hamada N, Wada S, Tanaka A, Shinagawa M,
Ohtsuki T, Mori T, Saha MN, Hoque AS, Islam S, Kogure K, Funayama T,
Kobayashi Y, Hoshino H (2010) Irradiation with carbon ion beams
induces apoptosis, autophagy, and cellular senescence in a human glioma-
derived cell line. Int J Radiat Oncol Biol Phys 76: 229–241.
Jo GH, Bo
¨gler O, Chwae YJ, Yoo H, Lee SH, Park JB, Kim YJ, Kim JH,
Gwak HS (2014) Radiation-induced autophagy contributes to cell death
and induces apoptosis partly in malignant glioma cells. Cancer Res 47:
221–241.
Kabeya Y, Mizushima N, Ueno T (2000) LC3, a mammalian homologue of
yeast Apg8p, is localized in autophagosome membranes after processing.
EMBO J 19: 5720–5728.
Kalamida D, Karagounis IV, Mitrakas A, Kalamida S, Giatromanolaki A,
Koukourakis MI (2015) Fever-range hyperthermia vs hypothermia effect
on cancer cell viability, proliferation and HSP90 expression. PLoS One 10:
e0116021.
BRITISH JOURNAL OF CANCER Autophagy, radiation and temozolomide
8 www.bjcancer.com | DOI:10.1038/bjc.2016.19
Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S (2004) Role of
autophagy in temozolomide-induced cytotoxicity for malignant glioma
cells. Cell Death Differ 11: 448–457.
Katayama M, Kawaguchi T, Berger MS, Pieper RO (2007) DNA damaging
agent-induced autophagy produces a cytoprotective adenosine
triphosphate surge in malignant glioma cells. Cell Death Differ 14:
548–558.
Katsuragi Y, Ichimura Y, Komatsu M (2015) p62/SQSTM1 functions as a
signaling hub and an autophagy adaptor. FEBS J 282: 4672–4678.
Kim H, Bernard ME, Flickinger J, Epperly MW, Wang H, Dixon TM,
Shields D, Houghton F, Zhang X, Greenberger JS (2011) The autophagy-
inducing drug carbamazepine is a radiation protector and mitigator. Int J
Radiat Biol 87: 1052–1060.
Kim SY, Yoo YH, Park JW (2013) Silencing of mitochondrial NADP( þ)-
dependent isocitrate dehydrogenase gene glioma radiosensitivity. Biochem
Biophys Res Commun 433: 260–265.
Kimura T, Takabatake Y, Takahashi A, Isaka Y (2013) Chloroquine in cancer
therapy: adouble-edged sword of autophagy. Cancer Res 73: 3–7.
Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A,
Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA, Ahn HJ,
Ait-Mohamed O, Ait-Si-Ali S, Akematsu T, Akira S, Al-Younes HM,
Al-Zeer MA, Albert ML, Albin RL, Alegre-Abarrategui J, Aleo MF,
Alirezaei M, Almasan A, Almonte-Becerril M, Amano A, Amaravadi R,
Amarnath S, Amer AO, Andrieu-Abadie N, Anantharam V, Ann DK,
Anoopkumar-Dukie S, Aoki H, Apostolova N, Arancia G, Aris JP,
Asanuma K, Asare NY, Ashida H, Askanas V, Askew DS, Auberger P,
Baba M, Backues SK, Baehrecke EH, Bahr BA, Bai XY, Bailly Y,
Baiocchi R, Baldini G, Balduini W, Ballabio A, Bamber BA, Bampton ET,
Ba
´nhegyi G, Bartholomew CR, Bassham DC, Bast Jr RC, Batoko H,
Bay BH, Beau I, Be
´chet DM, Begley TJ, Behl C, Behrends C, Bekri S,
Bellaire B, Bendall LJ, Benetti L, Berliocchi L, Bernardi H, Bernassola F,
Besteiro S, Bhatia-Kissova I, Bi X, Biard-Piechaczyk M, Blum JS, Boise LH,
Bonaldo P, Boone DL, Bornhauser BC, Bortoluci KR, Bossis I, Bost F,
Bourquin JP, Boya P, Boyer-Guittaut M, Bozhkov PV, Brady NR,
Brancolini C, Brech A, Brenman JE, Brennand A, Bresnick EH,
Brest P, Bridges D, Bristol ML, Brookes PS, Brown EJ, Brumell JH,
Brunetti-Pierri N, Brunk UT, Bulman DE, Bultman SJ, Bultynck G,
Burbulla LF, Bursch W, Butchar JP, Buzgariu W, Bydlowski SP, Cadwell K,
Cahova
´M, Cai D, Cai J, Cai Q, Calabretta B, Calvo-Garrido J,
Camougrand N, Campanella M, Campos-Salinas J, Candi E, Cao L,
Caplan AB, Carding SR, Cardoso SM, Carew JS, Carlin CR, Carmignac V,
Carneiro LA, Carra S, Caruso RA, Casari G, Casas C, Castino R,
Cebollero E, Cecconi F, Celli J, Chaachouay H, Chae HJ, Chai CY,
Chan DC, Chan EY, Chang RC, Che CM, Chen CC, Chen GC, Chen GQ,
Chen M, Chen Q, Chen SS, Chen W, Chen X, Chen X, Chen X, Chen YG,
Chen Y, Chen Y, Chen YJ, Chen Z, Cheng A, Cheng CH, Cheng Y,
Cheong H, Cheong JH, Cherry S, Chess-Williams R, Cheung ZH,
Chevet E, Chiang HL, Chiarelli R, Chiba T, Chin LS, Chiou SH,
Chisari FV, Cho CH, Cho DH, Choi AM, Choi D, Choi KS, Choi ME,
Chouaib S, Choubey D, Choubey V, Chu CT, Chuang TH, Chueh SH,
Chun T, Chwae YJ, Chye ML, Ciarcia R, Ciriolo MR, Clague MJ, Clark RS,
Clarke PG, Clarke R, Codogno P, Coller HA, Colombo MI, Comincini S,
Condello M, Condorelli F, Cookson MR, Coombs GH, Coppens I,
Corbalan R, Cossart P, Costelli P, Costes S, Coto-Montes A, Couve E,
Coxon FP, Cregg JM, Crespo JL, Cronje
´MJ, Cuervo AM, Cullen JJ,
Czaja MJ, D’Amelio M, Darfeuille-Michaud A, Davids LM, Davies FE,
De Felici M, de Groot JF, de Haan CA, De Martino L, De Milito A,
De Tata V, Debnath J, Degterev A, Dehay B, Delbridge LM, Demarchi F,
Deng YZ, Dengjel J, Dent P, Denton D, Deretic V, Desai SD, Devenish RJ,
Di Gioacchino M, Di Paolo G, Di Pietro C, Dı
´az-Araya G, Dı
´az-Laviada I,
Diaz-Meco MT, Diaz-Nido J, Dikic I, Dinesh-Kumar SP, Ding WX,
Distelhorst CW, Diwan A, Djavaheri-Mergny M, Dokudovskaya S,
Dong Z, Dorsey FC, Dosenko V, Dowling JJ, Doxsey S, Dreux M,
Drew ME, Duan Q, Duchosal MA, Duff K, Dugail I, Durbeej M,
Duszenko M, Edelstein CL, Edinger AL, Egea G, Eichinger L, Eissa NT,
Ekmekcioglu S, El-Deiry WS, Elazar Z, Elgendy M, Ellerby LM, Eng KE,
Engelbrecht AM, Engelender S, Erenpreisa J, Escalante R, Esclatine A,
Eskelinen EL, Espert L, Espina V, Fan H, Fan J, Fan QW, Fan Z, Fang S,
Fang Y, Fanto M, Fanzani A, Farkas T, Farre
´JC, Faure M, Fechheimer M,
Feng CG, Feng J, Feng Q, Feng Y, Fe
´su¨s L, Feuer R, Figueiredo-Pereira ME,
Fimia GM, Fingar DC, Finkbeiner S, Finkel T, Finley KD, Fiorito F,
Fisher EA, Fisher PB, Flajolet M, Florez-McClure ML, Florio S, Fon EA,
Fornai F, Fortunato F, Fotedar R, Fowler DH, Fox HS, Franco R,
Frankel LB, Fransen M, Fuentes JM, Fueyo J, Fujii J, Fujisaki K, Fujita E,
Fukuda M, Furukawa RH, Gaestel M, Gailly P, Gajewska M, Galliot B,
Galy V, Ganesh S, Ganetzky B, Ganley IG, Gao FB, Gao GF, Gao J,
Garcia L, Garcia-Manero G, Garcia-Marcos M, Garmyn M, Gartel AL,
Gatti E, Gautel M, Gawriluk TR, Gegg ME, Geng J, Germain M,
Gestwicki JE, Gewirtz DA, Ghavami S, Ghosh P, Giammarioli AM,
Giatromanolaki AN, Gibson SB, Gilkerson RW, Ginger ML, Ginsberg HN,
Golab J, Goligorsky MS, Golstein P, Gomez-Manzano C, Goncu E,
Gongora C, Gonzalez CD, Gonzalez R, Gonza
´lez-Este
´vez C,
Gonza
´lez-Polo RA, Gonzalez-Rey E, Gorbunov NV, Gorski S, Goruppi S,
Gottlieb RA, Gozuacik D, Granato GE, Grant GD, Green KN, Gregorc A,
Gros F, Grose C, Grunt TW, Gual P, Guan JL, Guan KL, Guichard SM,
Gukovskaya AS, Gukovsky I, Gunst J, Gustafsson AB, Halayko AJ,
Hale AN, Halonen SK, Hamasaki M, Han F, Han T, Hancock MK,
Hansen M, Harada H, Harada M, Hardt SE, Harper JW, Harris AL,
Harris J, Harris SD, Hashimoto M, Haspel JA, Hayashi S, Hazelhurst LA,
He C, He YW, He
´bert MJ, Heidenreich KA, Helfrich MH, Helgason GV,
Henske EP, Herman B, Herman PK, Hetz C, Hilfiker S, Hill JA,
Hocking LJ, Hofman P, Hofmann TG, Ho
¨hfeld J, Holyoake TL, Hong
MH, Hood DA, Hotamisligil GS, Houwerzijl EJ, Høyer-Hansen M, Hu B,
Hu CA, Hu HM, Hua Y, Huang C, Huang J, Huang S, Huang WP,
Huber TB, Huh WK, Hung TH, Hupp TR, Hur GM, Hurley JB,
Hussain SN, Hussey PJ, Hwang JJ, Hwang S, Ichihara A, Ilkhanizadeh S,
Inoki K, Into T, Iovane V, Iovanna JL, Ip NY, Isaka Y, Ishida H, Isidoro C,
Isobe K, Iwasaki A, Izquierdo M, Izumi Y, Jaakkola PM, Ja
¨a
¨ttela
¨M,
Jackson GR, Jackson WT, Janji B, Jendrach M, Jeon JH, Jeung EB, Jiang H,
Jiang H, Jiang JX, Jiang M, Jiang Q, Jiang X, Jiang X, Jime
´nez A, Jin M,
Jin S, Joe CO, Johansen T, Johnson DE, Johnson GV, Jones NL, Joseph B,
Joseph SK, Joubert AM, Juha
´sz G, Juillerat-Jeanneret L, Jung CH, Jung YK,
Kaarniranta K, Kaasik A, Kabuta T, Kadowaki M, Kagedal K, Kamada Y,
Kaminskyy VO, Kampinga HH, Kanamori H, Kang C, Kang KB, Kang KI,
Kang R, Kang YA, Kanki T, Kanneganti TD, Kanno H, Kanthasamy AG,
Kanthasamy A, Karantza V, Kaushal GP, Kaushik S, Kawazoe Y, Ke PY,
Kehrl JH, Kelekar A, Kerkhoff C, Kessel DH, Khalil H, Kiel JA, Kiger AA,
Kihara A, Kim DR, Kim DH, Kim DH, Kim EK, Kim HR, Kim JS, Kim JH,
Kim JC, Kim JK, Kim PK, Kim SW, Kim YS, Kim Y, Kimchi A,
Kimmelman AC, King JS, Kinsella TJ, Kirkin V, Kirshenbaum LA,
Kitamoto K, Kitazato K, Klein L, Klimecki WT, Klucken J, Knecht E,
Ko BC, Koch JC, Koga H, Koh JY, Koh YH, Koike M, Komatsu M,
Kominami E, Kong HJ, Kong WJ, Korolchuk VI, Kotake Y, Koukourakis MI,
Kouri Flores JB, Kova
´cs AL, Kraft C, Krainc D, Kra
¨mer H, Kretz-Remy C,
Krichevsky AM, Kroemer G, Kru¨ger R, Krut O, Ktistakis NT, Kuan CY,
Kucharczyk R, Kumar A, Kumar R, Kumar S, Kundu M, Kung HJ, Kurz T,
Kwon HJ, La Spada AR, Lafont F, Lamark T, Landry J, Lane JD,
Lapaquette P, Laporte JF, La
´szlo
´L, Lavandero S, Lavoie JN, Layfield R,
Lazo PA, Le W, Le Cam L, Ledbetter DJ, Lee AJ, Lee BW, Lee GM, Lee J,
Lee JH, Lee M, Lee MS, Lee SH, Leeuwenburgh C, Legembre P, Legouis R,
Lehmann M, Lei HY, Lei QY, Leib DA, Leiro J, Lemasters JJ, Lemoine A,
Lesniak MS, Lev D, Levenson VV, Levine B, Levy E, Li F, Li JL, Li L, Li S,
Li W, Li XJ, Li YB, Li YP, Liang C, Liang Q, Liao YF, Liberski PP,
Lieberman A, Lim HJ, Lim KL, Lim K, Lin CF, Lin FC, Lin J, Lin JD,
Lin K, Lin WW, Lin WC, Lin YL, Linden R, Lingor P, Lippincott-Schwartz J,
Lisanti MP, Liton PB, Liu B, Liu CF, Liu K, Liu L, Liu QA, Liu W, Liu YC,
Liu Y, Lockshin RA, Lok CN, Lonial S, Loos B, Lopez-Berestein G,
Lo
´pez-Otı
´n C, Lossi L, Lotze MT, L+
ow P, Lu B, Lu B, Lu B, Lu Z,
Luciano F, Lukacs NW, Lund AH, Lynch-Day MA, Ma Y, Macian F,
MacKeigan JP, Macleod KF, Madeo F, Maiuri L, Maiuri MC, Malagoli D,
Malicdan MC, Malorni W, Man N, Mandelkow EM, Manon S, Manov I,
Mao K, Mao X, Mao Z, Marambaud P, Marazziti D, Marcel YL,
Marchbank K, Marchetti P, Marciniak SJ, Marcondes M, Mardi M,
Marfe G, Marin
˜o G, Markaki M, Marten MR, Martin SJ, Martinand-Mari C,
Martinet W, Martinez-Vicente M, Masini M, Matarrese P, Matsuo S,
Matteoni R, Mayer A, Mazure NM, McConkey DJ, McConnell MJ,
McDermott C, McDonald C, McInerney GM, McKenna SL, McLaughlin B,
McLean PJ, McMaster CR, McQuibban GA, Meijer AJ, Meisler MH,
Mele
´ndez A, Melia TJ, Melino G, Mena MA, Menendez JA,
Menna-Barreto RF, Menon MB, Menzies FM, Mercer CA, Merighi A,
Merry DE, Meschini S, Meyer CG, Meyer TF, Miao CY, Miao JY,
Michels PA, Michiels C, Mijaljica D, Milojkovic A, Minucci S, Miracco C,
Miranti CK, Mitroulis I, Miyazawa K, Mizushima N, Mograbi B,
Mohseni S, Molero X, Mollereau B, Mollinedo F, Momoi T,
Monastyrska I, Monick MM, Monteiro MJ, Moore MN, Mora R,
Moreau K, Moreira PI, Moriyasu Y, Moscat J, Mostowy S, Mottram JC,
Autophagy, radiation and temozolomide BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2016.19 9
Motyl T, Moussa CE, Mu¨ller S, Muller S, Mu¨nger K, Mu¨nz C, Murphy LO,
Murphy ME, Musaro
`A, Mysorekar I, Nagata E, Nagata K, Nahimana A,
Nair U, Nakagawa T, Nakahira K, Nakano H, Nakatogawa H,
Nanjundan M, Naqvi NI, Narendra DP, Narita M, Navarro M,
Nawrocki ST, Nazarko TY, Nemchenko A, Netea MG, Neufeld TP,
Ney PA, Nezis IP, Nguyen HP, Nie D, Nishino I, Nislow C, Nixon RA,
Noda T, Noegel AA, Nogalska A, Noguchi S, Notterpek L, Novak I,
Nozaki T, Nukina N, Nu¨ rnberger T, Nyfeler B, Obara K, Oberley TD,
Oddo S, Ogawa M, Ohashi T, Okamoto K, Oleinick NL, Oliver FJ,
Olsen LJ, Olsson S, Opota O, Osborne TF, Ostrander GK, Otsu K, Ou JH,
Ouimet M, Overholtzer M, Ozpolat B, Paganetti P, Pagnini U, Pallet N,
Palmer GE, Palumbo C, Pan T, Panaretakis T, Pandey UB, Papackova Z,
Papassideri I, Paris I, Park J, Park OK, Parys JB, Parzych KR, Patschan S,
Patterson C, Pattingre S, Pawelek JM, Peng J, Perlmutter DH, Perrotta I,
Perry G, Pervaiz S, Peter M, Peters GJ, Petersen M, Petrovski G, Phang JM,
Piacentini M, Pierre P, Pierrefite-Carle V, Pierron G, Pinkas-Kramarski R,
Piras A, Piri N, Platanias LC, Po
¨ggeler S, Poirot M, Poletti A, Pou¨s C,
Pozuelo-Rubio M, Prætorius-Ibba M, Prasad A, Prescott M, Priault M,
Produit-Zengaffinen N, Progulske-Fox A, Proikas-Cezanne T,
Przedborski S, Przyklenk K, Puertollano R, Puyal J, Qian SB, Qin L,
Qin ZH, Quaggin SE, Raben N, Rabinowich H, Rabkin SW, Rahman I,
Rami A, Ramm G, Randall G, Randow F, Rao VA, Rathmell JC,
Ravikumar B, Ray SK, Reed BH, Reed JC, Reggiori F, Re
´gnier-Vigouroux A,
Reichert AS, Reiners Jr JJ, Reiter RJ, Ren J, Revuelta JL, Rhodes CJ, Ritis K,
Rizzo E, Robbins J, Roberge M, Roca H, Roccheri MC, Rocchi S,
Rodemann HP, Rodrı
´guez de Co
´rdoba S, Rohrer B, Roninson IB, Rosen K,
Rost-Roszkowska MM, Rouis M, Rouschop KM, Rovetta F, Rubin BP,
Rubinsztein DC, Ruckdeschel K, Rucker III EB, Rudich A, Rudolf E,
Ruiz-Opazo N, Russo R, Rusten TE, Ryan KM, Ryter SW, Sabatini DM,
Sadoshima J, Saha T, Saitoh T, Sakagami H, Sakai Y, Salekdeh GH,
Salomoni P, Salvaterra PM, Salvesen G, Salvioli R, Sanchez AM,
Sa
´nchez-Alca
´zar JA, Sa
´nchez-Prieto R, Sandri M, Sankar U, Sansanwal P,
Santambrogio L, Saran S, Sarkar S, Sarwal M, Sasakawa C, Sasnauskiene A,
Sass M, Sato K, Sato M, Schapira AH, Scharl M, Scha
¨tzl HM, Scheper W,
Schiaffino S, Schneider C, Schneider ME, Schneider-Stock R, Schoenlein PV,
Schorderet DF, Schu¨ller C, Schwartz GK, Scorrano L, Sealy L, Seglen PO,
Segura-Aguilar J, Seiliez I, Seleverstov O, Sell C, Seo JB, Separovic D,
Setaluri V, Setoguchi T, Settembre C, Shacka JJ, Shanmugam M,
Shapiro IM, Shaulian E, Shaw RJ, Shelhamer JH, Shen HM, Shen WC,
Sheng ZH, Shi Y, Shibuya K, Shidoji Y, Shieh JJ, Shih CM, Shimada Y,
Shimizu S, Shintani T, Shirihai OS, Shore GC, Sibirny AA, Sidhu SB,
Sikorska B, Silva-Zacarin EC, Simmons A, Simon AK, Simon HU,
Simone C, Simonsen A, Sinclair DA, Singh R, Sinha D, Sinicrope FA,
Sirko A, Siu PM, Sivridis E, Skop V, Skulachev VP, Slack RS, Smaili SS,
Smith DR, Soengas MS, Soldati T, Song X, Sood AK, Soong TW, Sotgia F,
Spector SA, Spies CD, Springer W, Srinivasula SM, Stefanis L, Steffan JS,
Stendel R, Stenmark H, Stephanou A, Stern ST, Sternberg C, Stork B,
Strålfors P, Subauste CS, Sui X, Sulzer D, Sun J, Sun SY, Sun ZJ, Sung JJ,
Suzuki K, Suzuki T, Swanson MS, Swanton C, Sweeney ST, Sy LK,
Szabadkai G, Tabas I, Taegtmeyer H, Tafani M, Taka
´cs-Vellai K,
Takano Y, Takegawa K, Takemura G, Takeshita F, Talbot NJ, Tan KS,
Tanaka K, Tanaka K, Tang D, Tang D, Tanida I, Tannous BA,
Tavernarakis N, Taylor GS, Taylor GA, Taylor JP, Terada LS, Terman A,
Tettamanti G, Thevissen K, Thompson CB, Thorburn A, Thumm M,
Tian F, Tian Y, Tocchini-Valentini G, Tolkovsky AM, Tomino Y,
To
¨nges L, Tooze SA, Tournier C, Tower J, Towns R, Trajkovic V,
Travassos LH, Tsai TF, Tschan MP, Tsubata T, Tsung A, Turk B,
Turner LS, Tyagi SC, Uchiyama Y, Ueno T, Umekawa M,
Umemiya-Shirafuji R, Unni VK, Vaccaro MI, Valente EM, Van den Berghe G,
van der Klei IJ, van Doorn W, van Dyk LF, van Egmond M, van Grunsven LA,
Vandenabeele P, Vandenberghe WP, Vanhorebeek I, Vaquero EC,
Velasco G, Vellai T, Vicencio JM, Vierstra RD, Vila M, Vindis C, Viola G,
Viscomi MT, Voitsekhovskaja OV, von Haefen C, Votruba M, Wada K,
Wade-Martins R, Walker CL, Walsh CM, Walter J, Wan XB, Wang A,
Wang C, Wang D, Wang F, Wang F, Wang G, Wang H, Wang HG,
Wang HD, Wang J, Wang K, Wang M, Wang RC, Wang X, Wang X,
Wang YJ, Wang Y, Wang Z, Wang ZC, Wang Z, Wansink DG, Ward DM,
Watada H, Waters SL, Webster P, Wei L, Weihl CC, Weiss WA,
Welford SM, Wen LP, Whitehouse CA, Whitton JL, Whitworth AJ,
Wileman T, Wiley JW, Wilkinson S, Willbold D, Williams RL,
Williamson PR, Wouters BG, Wu C, Wu DC, Wu WK, Wyttenbach A,
Xavier RJ, Xi Z, Xia P, Xiao G, Xie Z, Xie Z, Xu DZ, Xu J, Xu L, Xu X,
Yamamoto A, Yamamoto A, Yamashina S, Yamashita M, Yan X,
Yanagida M, Yang DS, Yang E, Yang JM, Yang SY, Yang W, Yang WY,
Yang Z, Yao MC, Yao TP, Yeganeh B, Yen WL, Yin JJ, Yin XM, Yoo OJ,
Yoon G, Yoon SY, Yorimitsu T, Yoshikawa Y, Yoshimori T, Yoshimoto K,
You HJ, Youle RJ, Younes A, Yu L, Yu L, Yu SW, Yu WH, Yuan ZM,
Yue Z, Yun CH, Yuzaki M, Zabirnyk O, Silva-Zacarin E, Zacks D,
Zacksenhaus E, Zaffaroni N, Zakeri Z, Zeh III HJ, Zeitlin SO, Zhang H,
Zhang HL, Zhang J, Zhang JP, Zhang L, Zhang L, Zhang MY, Zhang XD,
Zhao M, Zhao YF, Zhao Y, Zhao ZJ, Zheng X, Zhivotovsky B, Zhong Q,
Zhou CZ, Zhu C, Zhu WG, Zhu XF, Zhu X, Zhu Y, Zoladek T, Zong WX,
Zorzano A, Zschocke J, Zuckerbraun B (2012) Guidelines for the use and
interpretation of assays for monitoring autophagy. Autophagy 8: 445–544.
Klionsky DJ, Emr SD (2000) Autophagy as a regulated pathway of cellular
degradation. Science 290: 1717–1721.
Koritzinsky M, Wouters BG (2013) The roles of reactive oxygen species and
autophagy in mediating the tolerance of tumor cells to cycling hypoxia.
Semin Radiat Oncol 23: 252–261.
Koukourakis MI, Kalamida D, Giatromanolaki A, Zois C, Sivridis E,
Pouliliou S, Mitrakas A, Gatter GC, Harris AL (2015) Autophagosome
proteins LC3A, LC3B and LC3C have distinct subcellular distribution
kinetics and expression in cancer cell lines. PLoS One 10(9): e0137675.
Koukourakis MI, Kalamida D, Mitrakas A, Pouliliou S, Kalamida S, Sivridis E,
Giatromanolaki A (2015) Intensified autophagy compromises the efficacy
of radiotherapy against prostate cancer. Biochem Biophys Res Commun
461: 268–274.
Krakstad C, Chekenya M (2010) Survival signalling and apoptosis resistance
in glioblastomas: opportunities for targeted therapeutics. Mol Cancer
9: 135.
Kreisl TN, Kim L, Moore K (2009) Phase II trial of single-agent bevacizumab
followed by bevacizumab plus irinotecan at tumour progression in
recurrent glioblastoma. J Clin Oncol 27: 740–745.
Lee JJ, Kim BC, Park MJ, Lee YS, Kim YN, Lee BL, Lee JS (2011) PTEN status
switches cell fate between premature senescence and apoptosis in glioma
exposed to ionizing radiation. Cell Death Differ 18: 666–677.
Lee SW, Kim HK, Lee NH, Yi HY, Kim HS, Hong SH, Hong YK, Joe YA
(2015) The synergistic effect of combination temozolomide and
chloroquine treatment is dependent on autophagy formation and p53
status in glioma cells. Cancer Lett 360: 195–204.
Lefranc F, Kiss R (2006) Autophagy, the Trojan horse to combat
glioblastomas. Neurosurg Focus 20: E7.
Lena A, Rechichi M, Salvetti A, Bartoli B, Vecchio D, Scarcelli V, Amoroso R,
Benvenuti L, Gagliardi R, Gremigni V, Rossi L (2009) Drugs targeting the
mitochondrial pore act as cytotoxic and cytostatic agents in
temozolomide-resistant glioma cells. J Transl Med 7: 13.
Li Z, Chen B, Wu Y, Jin F, Xia Y, Liu X (2010) Genetic and epigenetic silencing
of the beclin 1 gene in sporadic breast tumours. BMC Cancer 10: 98.
Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN,
Mikkelsen T, Brodie C (2009) The induction of autophagy by gamma-
radiation contributes to the radioresistance of glioma stem cells. Int J
Cancer 125: 717–722.
Ma DJ, Galanis E, Anderson SK, Schiff D, Kaufmann TJ, Peller PJ, Giannini C,
Brown PD, Uhm JH, McGraw S, Jaeckle KA, Flynn PJ, Ligon KL,
Buckner JC, Sarkaria JN (2015) A phase II trial of everolimus,
temozolomide, and radiotherapy in patients with newly diagnosed
glioblastoma: NCCTG N057K. Neuro-Oncology 17: 1261–1269.
Martelli AM, Evangelisti C, Follo MY, Ramazzotti G, Fini M, Giardino R,
Manzoli L, McCubrey JA, Cocco L (2011) Targeting the
phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin
signaling network in cancer stem cells. Curr Med Chem 18: 2715–2726.
Mehta M, Khan A, Danish S, Haffty BG, Sabaawy HE (2015)
Radiosensitization of primary human glioblastoma stem-like cells with
low-dose AKT inhibition. Mol Cancer Ther 14: 1171–1180.
Miao W, Liu X, Wang H, Fan Y, Lian S, Yang X, Wang X, Guo G, Li Q,
Wang S (2015) p53 upregulated modulator of apoptosis sensitizes drug-
resistant U251 glioblastoma stem cells to temozolomide through enhanced
apoptosis. Mol Med Rep 11: 4165–4173.
Milano V, Piao Y, LaFortune T, de Groot J (2009) Dasatinib-induced
autophagy is enhanced in combination with temozolomide in glioma. Mol
Cancer Ther 8: 394–406.
Mirzayans R, Andrais B, Scott A, Wang YW, Murray D (2013) Ionizing
radiation-induced responses in human cells with differing TP53 status. Int
J Mol Sci 14: 22409–22435.
BRITISH JOURNAL OF CANCER Autophagy, radiation and temozolomide
10 www.bjcancer.com | DOI:10.1038/bjc.2016.19
Morgan MA, Lawrence TS (2015) Molecular pathways: overcoming radiation
resistance by targeting DNA damage response pathways. Clin Cancer Res
21: 2898–28904.
Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL,
Burkhard C, Schu¨ ler D, Probst-Hensch NM, Maiorka PC, Baeza N,
Pisani P, Yonekawa Y, Yasargil MG, Lu¨tolf UM, Kleihues P (2004) Genetic
pathways to glioblastoma: a population-based study. Cancer Res 64:
6892–6899.
Palumbo S, Tini P, Toscano M, Allavena G, Angeletti F, Manai F, Miracco C,
Comincini S, Pirtoli L (2014) Combined EGFR and autophagy modulation
impairs cell migration and enhances radiosensitivity in human
glioblastoma cells. J Cell Physiol 229: 1863–1873.
Park JA, Kim YE, Seok HJ, Park WY, Kwon HJ, Lee Y (2011) Differentiation
and upregulation of heat shock protein 70 induced by a subset of histone
deacetylase inhibitors in mouse and human embryonic stem cells. BMB
Rep 44: 176–181.
Pellegrini P, Strambi A, Zipoli C, Ha
¨gg-Olofsson M, Buoncervello M,
Linder S, De Milito A (2014) Acidic extracellular pH neutralizes the
autophagy-inhibiting activity of chloroquine: implications for cancer
therapies. Autophagy 10: 562–571.
Pflaum J, Schlosser S, Mu¨ller M (2014) p53 Family and cellular stress
responses in cancer. Front Oncol 4: 285.
Pirtoli L, Cevenini G, Tini P, Vannini M, Oliveri G, Marsili S, Mourmouras V,
Rubino G, Miracco C (2009) The prognostic role of Beclin 1 protein
expression in high-grade gliomas. Autophagy 5: 930–936.
Poulsen HS, Urup T, Michaelsen SR, Staberg M, Villingshøj M, Lassen U
(2014) The impact of bevacizumab treatment on survival and quality of
life in newly diagnosed glioblastoma patients. Cancer Manag Res 6:
373–387.
Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S,
Mikkelson T, Wang D, Chang YC, Hu J, McAfee Q, Fisher J, Troxel AB,
Piao S, Heitjan DF, Tan KS, Pontiggia L, O’Dwyer PJ, Davis LE,
Amaravadi RK (2014) A phase I/II trial of hydroxychloroquine in
conjunction with radiation therapy and concurrent and adjuvant
temozolomide in patients with newly diagnosed glioblastoma multiforme.
Autophagy 10: 1359–1368.
Saetre F, Hagen LK, Engedal N, Seglen PO (2015) Novel steps in the
autophagic-lysosomal pathway. FEBS J 282: 2202–2214.
Salazar OM, Rubin P, Feldstein ML, Pizzutiello R (1979) High dose radiation
therapy in the treatment of malignant gliomas: final report. Int J Radiat
Oncol Biol Phys 5: 1733–1740.
Sarkaria JN, Galanis E, Wu W, Peller PJ, Giannini C, Brown PD, Uhm JH,
McGraw S, Jaeckle KA, Buckner JC (2011) North Central Cancer
Treatment Group Phase I trial N057K of everolimus (RAD001) and
temozolomide in combination with radiation therapy in patients with
newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys 81:
468–475.
Sesen J, Dahan P, Scotland SJ, Saland E, Dang VT, Lemarie
´A, Tyler BM,
Brem H, Toulas C, Cohen-Jonathan Moyal E, Sarry JE, Skuli N (2015)
Metformin inhibits growth of human glioblastoma cells and enhances
therapeutic response. PLoS One 10: e0123721.
Shapiro WR, Green SB, Burger PC (1989) Randomized trial of three
chemotherapy regimens and two radiotherapy regimens and two
radiotherapy regimens in postoperative treatment of malignant glioma.
Brain Tumour Cooperative Group Trial 8001. J Neurosurg 71: 1–9.
Shen Y, Chen H, Zhang J, Chen Y, Wang M, Ma J, Hong L, Liu N, Fan Q,
Lu X, Tian Y, Wang A, Dong J, Lan Q, Huang Q (2015) Increased Notch
signaling enhances radioresistance of malignant stromal cells induced by
glioma stem/progenitor cells. PLoS One 10(11): e0142594.
Shpilka T, Weidberg H, Pietrokovski S, Elazar Z (2011) Atg8: an autophagy-
related ubiquitin-like protein family. Genome Biol 12: 226.
Sivridis E, Koukourakis MI, Zois CE, Ledaki I, Ferguson DJ, Harris AL,
Gatter KC, Giatromanolaki A (2010) LC3A-positive light microscopy
detected patterns of autophagy and prognosis in operable breast
carcinomas. Am J Pathol 176: 2477–2489.
Stupp R (2009) European Organisation for Research and Treatment of Cancer
Brain Tumour and Radiation Oncology Groups; National Cancer Institute
of Canada Clinical Trials Group. Effects of radiotherapy with concomitant
and adjuvant temozolomide versus radiotherapy alone on survival in
glioblastoma in a randomised phase III study: 5-year analysis of the
EORTC-NCIC trial. Lancet Oncol 10: 459–466.
Stupp R, Mason WP, van den Bent MJ (2005) European Organisation for
Research and Treatment of Cancer Brain Tumour and Radiotherapy
Groups; National Cancer Institute of Canada Clinical Trials Group.
Radiotherapy plus concomitant and adjuvant temozolomide for
glioblastoma. N Engl J Med 352: 987–996.
Thomas AA, Omuro A (2014) Current role of anti-angiogenic strategies for
glioblastoma. Curr Treat Options Oncol 15: 551–566.
Tsuboi Y, Kurimoto M, Nagai S, Hayakawa Y, Kamiyama H, Hayashi N,
Kitajima I, Endo S (2009) Induction of autophagic cell death and
radiosensitization by the pharmacological inhibition of nuclear factor-
kappa B activation in human glioma cell lines. J Neurosurg 110: 594–604.
Tsurushima H, Yuan X, Dillehay LE, Leong KW (2008) Radiation-inducible
caspase-8 gene therapy for malignant brain tumours. Int J Radiat Oncol
Biol Phys 71: 517–525.
Ulasov IV, Sonabend AM, Nandi S, Khramtsov A, Han Y, Lesniak MS (2009)
Combination of adenoviral virotherapy and temozolomide chemotherapy
eradicates malignant glioma through autophagic and apoptotic cell death
in vivo. Br J Cancer 100: 1154–1164.
Vakifahmetoglu H, Olsson M, Zhivotovsky B (2008) Death through a tragedy:
mitotic catastrophe. Cell Death Differ 15: 1153–1162.
van den Bent MJ, Brandes AA, Rampling R, Kouwenhoven MC, Kros JM,
Carpentier AF, Clement PM, Frenay M, Campone M, Baurain JF,
Armand JP, Taphoorn MJ, Tosoni A, Kletzl H, Klughammer B,
Lacombe D, Gorlia T (2009) Randomized phase II trial of erlotinib versus
temozolomide or carmustine in recurrent glioblastoma: EORTC brain
tumour group study 26034. J Clin Oncol 27: 1268–1274.
Villalonga-Planells R, Coll-Mulet L, Martı
´nez-Soler F, Castan
˜o E, Acebes J,
Gime
´nez-Bonafe
´P, Gil J, Tortosa A (2011) Activation of p53 by nutlin-3a
induces apoptosis and cellular senescence in human glioblastoma
multiforme. PLoS One 6(4): e18588.10.
Winardi D, Tsai HP, Chai CY, Chung CL, Loh JK, Chen YH, Hsieh CL (2014)
Correlation of altered expression of the autophagy marker LC3B with poor
prognosis in astrocytoma. Biomed Res Int 2014: 723176.
Wong PM, Puente C, Ganley IG, Jiang X (2013) The ULK1 complex: sensing
nutrient signals for autophagy activation. Autophagy 9: 124–137.
Yacoub A, Hamed H, Emdad L, Dos Santos W, Gupta P, Broaddus WC,
Ramakrishnan V, Sarkar D, Shah K, Curiel DT, Grant S, Fisher PB, Dent P
(2008) MDA-7/IL-24 plus radiation enhance survival in animals with
intracranial primary human GBM tumours. Cancer Biol Ther 7: 917–933.
Yao KC, Komata T, Kondo Y (2003) Molecular response of human
glioblastoma multiforme cells to ionizing radiation: cell cycle arrest,
modulation of the expression of cyclin-dependent kinase inhibitors, and
autophagy. J Neurosurg 98: 378–384.
Yao KC, Komata T, Kondo Y, Kanzawa T, Kondo S, Germano IM (2003)
Molecular response of human glioblastoma multiforme cells to ionizing
radiation: cell cycle arrest, modulation of the expression of cyclin-
dependent kinase inhibitors, and autophagy. J Neurosurg 98: 378–384.
Ye F, Zhang Y, Liu Y, Yamada K, Tso JL, Menjivar JC, Tian JY, Yong WH,
Schaue D, Mischel PS, Cloughesy TF, Nelson SF, Liau LM, McBride W,
Tso CL (2013) Protective properties of radio chemoresistant glioblastoma
stem cell clones are associated with metabolic adaptation to reduced
glucose dependence. PLoS One 8: e80397.
Yokoyama T, Iwado E, Kondo Y, Aoki H, Hayashi Y, Georgescu MM,
Sawaya R, Hess KR, Mills GB, Kawamura H, Hashimoto Y, Urata Y,
Fujiwara T, Kondo S (2008) Autophagy-inducing agents augment the
antitumour effect of telerase-selve oncolytic adenovirus OBP-405 on
glioblastoma cells. Gene Therapy 15: 1233–1239.
Yorimitsu T, Klionsky DJ (2005) Autophagy: molecular machinery for self
eating. Cell Death Differ. 2: 1542–1552.
You WC, Chiou SH, Huang CY, Chiang SF, Yang CL, Sudhakar JN, Lin TY,
Chiang IP, Shen CC, Cheng WY, Lin JC, Shieh SH, Chow KC (2013)
Mitochondrial protein ATPase family, AAA domain containing 3A correlates
with radioresistance in glioblastoma. Neuro-Oncology 15: 1342–1352.
Yu VY, Nguyen D, Pajonk F, Kupelian P, Kaprealian T, Selch M, Low DA,
Sheng K (2015) Incorporating cancer stem cells in radiation therapy
treatment response modeling and the implication in glioblastoma
multiforme treatment resistance. Int J Radiat Oncol Biol Phys 91: 866–875.
Yuan X, Du J, Hua S, Zhang H, Gu C, Wang J, Yang L, Huang J, Yu J, Liu F
(2015) Suppression of autophagy augments the radiosensitizing effects of
STAT3 inhibition on human glioma cells. Exp Cell Res 330: 267–276.
Zanotto-Filho A, Braganhol E, Klafke K, Figueiro
´F, Terra SR, Paludo FJ,
Morrone M, Bristot IJ, Battastini AM, Forcelini CM, Bishop AJ,
Gelain DP, Moreira JC (2015) Autophagy inhibition improves the efficacy
of curcumin/temozolomide combination therapy in glioblastomas. Cancer
Lett 358: 220–231.
Autophagy, radiation and temozolomide BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2016.19 11
Zhivotovsky B, Joseph B, Orrenius S (1999) Tumour radiosensitivity and
apoptosis. Exp Cell Res 248: 10–17.
Zhuang W, Li B, Long L, Chen L, Huang Q, Liang Z (2011a) Induction of
autophagy promotes differentiation of glioma-initiating cells and their
radiosensitivity. Int J Cancer 129: 2720–2731.
Zhuang W, Li B, Long L, Chen L, Huang Q, Liang ZQ (2011b) Knockdown of
the DNA-dependent protein kinase catalytic subunit radiosensitizes
glioma-initiating cells by inducing autophagy. Brain Res 1371: 7–15.
Ziegler DS, Wright RD, Kesari S, Lemieux ME, Tran MA, Jain M, Zawel L,
Kung AL (2008) Resistance of human glioblastoma multiforme cells to
growth factor inhibitors is overcome by blockade of inhibitor of apoptosis
proteins. J Clin Invest 118: 3109–3122.
Zois CE, Giatromanolaki A, Kainulainen H, Botaitis S, Torvinen S,
Simopoulos C, Kortsaris A, Sivridis E, Koukourakis MI (2011) Lung
autophagic response following exposure of mice to whole body irradiation,
with and without amifostine. Biochem Biophys Res Commun 404: 552–558.
Zois CE, Giatromanolaki A, Sivridis E, Papaiakovou M, Kainulainen H,
Koukourakis MI (2011) ‘Autophagic flux’ in normal mouse tissues: focus
on endogenous LC3A processing. Autophagy 7: 1371–1378.
Zois CE, Koukourakis MI (2009) Radiation-induced autophagy in normal and
cancer cells: towards novel cytoprotection and radio-sensitization policies?
Autophagy 5: 442–450.
This work is licensed under the Creative Commons
Attribution-Non-Commercial-Share Alike 4.0 Inter-
national License. To view a copy of this license, visit http://
creativecommons.org/licenses/by-nc-sa/4.0/
BRITISH JOURNAL OF CANCER Autophagy, radiation and temozolomide
12 www.bjcancer.com | DOI:10.1038/bjc.2016.19
... To ascertain the effectiveness, doses of 0.5 mM, 0.75 mM, 1 mM, and 1.25 mM TMZ and doses of 1 nM, 5 nM, 10 nM, 15 nM, and 20 nM ERB were applied to the T98G cell line, taking previous studies into consideration, 18,19 while the control group received no treatment. Dimethyl sulfoxide (DMSO) was used as a solvent for the drugs, therefore, an equivalent DMSO was applied to the DMSO group to make sure that the effects are not related to the DMSO. ...
Article
Full-text available
Glioblastoma tumors are the most aggressive primary brain tumors that develop resistance to temozolomide (TMZ). Eribulin (ERB) exhibits a unique mechanism of action by inhibiting microtubule dynamics during the G2/M cell cycle phase. We utilized the T98G human glioma cell line to investigate the effects of ERB and TMZ, both individually and in combination. The experimental groups were established as follows: control, E5 (5 nM ERB), T0.75 (0.75 mM TMZ), T1 (1.0 mM TMZ), and combination groups (E5+T0.75 and E5+T1). All groups showed a significant decrease in cell proliferation. Apoptotic markers revealed a time-dependent increase in annexin-V expression, across all treatment groups at the 48-hour time point. Caspase-3, exhibited an increase in the combination treatment groups at the 48-hour mark. Transmission electron microscopy (TEM) revealed normal ultrastructural features in the glioma cells of the control group. However, treatments induced ultrastructural changes within the spheroid glioblastoma model, particularly in the combination groups. These changes included a dose-dependent increase in autophagic vacuoles and apoptotic morphology of the cells. In conclusion, the similarity in the mechanism of action between ERB and TMZ suggests the potential for synergistic effects when combined. Our results highlight that this combination induced severe damage and autophagy in glioma spheroids after 48 hours. Keywords: Apoptosis; Eribulin; T98G; TEM; Temozolomide.
... We identified NUPR1 as one of potential mediators of GBM cell resistance to killing by TMZ and irradiation in CSF. TMZ and irradiation are classically known to kill cancer cells by causing DNA damage, inadequate DNA repair, cell cycle arrest, apoptosis, autophagy, and necrosis (56,57). Recently, TMZ and irradiation have also been reported to induce ferroptosis (58,59), a form of cell death triggered by altered redox homeostasis, iron overload, and increased lipid peroxidation (60). ...
Article
Full-text available
Cancers in the central nervous system resist therapies effective in other cancers, possibly due to the unique biochemistry of the human brain microenvironment composed of cerebrospinal fluid (CSF). However, the impact of CSF on cancer cells and therapeutic efficacy is unknown. Here, we examined the effect of human CSF on glioblastoma (GBM) tumors from 25 patients. We found that CSF induces tumor cell plasticity and resistance to standard GBM treatments (temozolomide and irradiation). We identified nuclear protein 1 (NUPR1), a transcription factor hampering ferroptosis, as a mediator of therapeutic resistance in CSF. NUPR1 inhibition with a repurposed antipsychotic, trifluoperazine, enhanced the killing of GBM cells resistant to chemoradiation in CSF. The same chemo-effective doses of trifluoperazine were safe for human neurons and astrocytes derived from pluripotent stem cells. These findings reveal that chemoradiation efficacy decreases in human CSF and suggest that combining trifluoperazine with standard care may improve the survival of patients with GBM.
... LC3 is an autophagosome marker 56 ; p62/SQSTM1/A170 is an autophagy-adaptor protein with an important role in directing and compacting ubiquitinated cargos towards autophagy 57 ; and ATG5 is considered to be essential for the induction of autophagy 58 . During stressful conditions, autophagy is critical for the maintenance of cellular homeostasis 59 , regulating the pro-growth signaling cascade and activating the metabolic rewiring of tumor cells to support their growth even in nutrientdeprived microenvironments 60,61 . Then, when the proteostasis by the UPS is perturbed, Nt-Arg redirects the cellular wastes to macroautophagy through p62 self-polymerization upon its binding to the Nt-Arg, which facilitates cargo collection and lysosomal degradation of p62 cargo-complexes. ...
Preprint
Full-text available
Post-translational modifications (PTM) have been recognized as a relevant regulation of key processes in cancer pathophysiology, such as cell migration, adhesion, and proliferation. N-terminal protein arginylation is an emerging PTM involved in tumor progression; however, the mechanisms by which this modification influences these events are poorly understood and vary according to cancer type. Glioblastoma (GBM) is an aggressive intra-axial brain tumor associated with poor prognosis, low survival, and high recurrence rate. We performed a study combining in silico, in vitro, and patients samples analysis to understand the impact of N-terminal protein arginylation in GBM, including overexpression and silencing of ATE1 in GBM-U87MG cell line with RNASeq analysis, immunofluorescence, and validation of the identified targets at the protein level by immunoblotting. The arginylation pattern differed in GBM compared with non-neoplastic brain tissues, and upregulation of ATE1 was associated with increased tumor cell proliferation. We identified a strong activation of the unfolded protein response (UPR) pathway associated with increased ATE1 level, inducing autophagy and not apoptosis. Protein arginylation in GBM proved to be an important mechanism for tumor growth, with the recycling of cell substrates by autophagy, providing fitness for tumor cells. The expressions of the main markers of UPR and autophagy pathways were validated in human GBM samples, reinforcing the role of ATE1 in the most aggressive brain tumor.
... Consequently, akin to early phase inhibitors, they induce cell death in a variable proportion of tumor cells by depriving them of energy and overloading them with waste. 34 Moreover, the coadministration of autophagy flux inhibitors with autophagy induction therapy, such as radiotherapy or TMZ, can intensify the accumulation of autophagosomes, leading to a superadditive effect. Notably, the use of autophagy inhibitors, such as CQ or HCQ, to impede the autophagy response has been demonstrated to enhance the sensitivity of tumor cells to cytotoxic therapy. ...
Article
Full-text available
Gliomas, the most lethal brain tumors, often exhibit resistance to conventional chemotherapy and/or radiotherapy. This study reveals that sertindole, a potent dopamine D2 receptor antagonist primarily designed as an antipsychotic medication for schizophrenia, effectively inhibits glioma progression. Our findings demonstrate that sertindole suppresses the proliferation of U251 and U87 tumor cells, impedes cell cycle progression in vitro, and curtails xenograft tumor growth in vivo. Moreover, we present compelling evidence demonstrating the ability of sertindole to enhance the cellular response to the chemotherapeutic agent temozolomide both in vitro and in vivo. Additionally, our findings reveal that sertindole effectively suppresses the self‐renewal capacity and expression of stemness‐associated genes, such as Nanog and Sox2, in glioma tumor cells and glioma stem cells. A mechanistic investigation demonstrated that sertindole enhances the formation of autophagosome–lysosome complexes while concurrently impeding autophagic flux through the inhibition of lysosomal hydrolytic enzymes CTSD and CTSB, ultimately resulting in decreased growth of tumor cells. In conclusion, our findings suggest that sertindole has the potential to develop into a potent antiglioma therapeutic agent.
... TMZ), the intracellular ADP and AMP concentration increases, activating AMP-dependent protein kinase (AMPK)-ULK1-Beclin1 pathway, which in turn induces autophagy and promotes cell survival [42]. Exposure of GBM cells to TMZ caused activation of AMPK and an increase in LC3-positive autophagosomes [43]. The autophagy inhibitors Baf and CQ promoted TMZ-induced apoptosis in glioma cells by preventing the fusion of mature autophagosomes with lysosomes [44]. ...
Article
Full-text available
The standard regimen treatment has improved GBM outcomes, but the survival rate of patients is still unsatisfactory. Temozolomide (TMZ) resistance is one of main reasons limiting the therapeutic efficacy of GBM. However, there are currently no TMZ-sensitizing drugs available in the clinic. Here we aimed to study whether the antidiabetic drug Sitagliptin can inhibit the survival, stemness and autophagy of GBM cells, and thus enhance TMZ cytotoxicity. We used CCK-8, EdU, colony formation, TUNEL and flow cytometry assays to assess cell proliferation and apoptosis; sphere formation and limiting dilution assays to measure self-renewal and stemness of glioma stem cells (GSCs); Western blot, qRT-PCR or immunohistochemical analysis to measure the expression of proliferation or stem cell markers; Western blot/fluorescent analysis of LC3 and other molecules to evaluate autophagy formation and degradation in glioma cells. We found that Sitagliptin inhibited proliferation and induced apoptosis in GBM cells and suppressed self-renewal and stemness of GSCs. The in vitro findings were further confirmed in glioma intracranial xenograft models. Sitagliptin administration prolonged the survival time of tumor-bearing mice. Sitagliptin could inhibit TMZ-induced protective autophagy and enhance the cytotoxicity of TMZ in glioma cells. In addition, Sitagliptin acted as a dipeptidyl peptidase 4 inhibitor in glioma as well as in diabetes, but it did not affect the blood glucose level and body weight of mice. These findings suggest that Sitagliptin with established pharmacologic and safety profiles could be repurposed as an antiglioma drug to overcome TMZ resistance, providing a new option for GBM therapy.
... TMZ has demonstrated an ability to upregulate autophagy in GBM both in vitro and in vivo [41,44]. Suppressing the phosphoinositide 3-kinase (PI3K) and protein kinase B (also known as Akt) axis while upregulating the mitogen-activated protein kinase (MAPK) pathway can lead to enhancement of autophagy activation following TMZ administration [45]. An increase in activation of autophagy has been tracked on the molecular levels, showing statistically significant changes in the microtubule-associated protein 1 light chain 3 isoform B (colloquially called LC3B), lysosome-associated membrane protein 1 (LAMP1), and LAMP2A following treatment with TMZ, indicating an upregulation of cytoprotective autophagy in patient samples. ...
Article
Full-text available
As the most malignant primary brain tumor in adults, a diagnosis of glioblastoma multiforme (GBM) continues to carry a poor prognosis. GBM is characterized by cytoprotective homeostatic processes such as the activation of autophagy, capability to confer therapeutic resistance, evasion of apoptosis, and survival strategy even in the hypoxic and nutrient-deprived tumor microenvironment. The current gold standard of therapy, which involves radiotherapy and concomitant and adjuvant chemotherapy with temozolomide (TMZ), has been a game-changer for patients with GBM, relatively improving both overall survival (OS) and progression-free survival (PFS); however, TMZ is now well-known to upregulate undesirable cytoprotective autophagy, limiting its therapeutic efficacy for induction of apoptosis in GBM cells. The identification of targets utilizing bioinformatics-driven approaches, advancement of modern molecular biology technologies such as clustered regularly interspaced short palindromic repeats (CRISPR)—CRISPR-associated protein (Cas9) or CRISPR-Cas9 genome editing, and usage of microRNA (miRNA)-mediated regulation of gene expression led to the selection of many novel targets for new therapeutic development and the creation of promising combination therapies. This review explores the current state of advanced bioinformatics analysis and genetic technologies and their utilization for synergistic combination with TMZ in the context of inhibition of autophagy for controlling the growth of GBM.
Article
Glioblastoma (GBM) is the most common malignant brain tumor and has a dismal prognosis even under the current first-line treatment, with a 5-year survival rate less than 7%. Therefore, it is important to understand the mechanism of treatment resistance and develop new anti-tumor strategies. Induction of programmed cell death (PCD) has become a promising anti-tumor strategy, but its effectiveness in treating GBM remains controversial. On the one hand, PCD triggers tumor cell death and then release mediators to draw in immune cells, creating a pro-inflammatory tumor microenvironment (TME). One the other hand, mounting evidence suggests that PCD and inflammatory TME will force tumor cells to evolve under survival stress, leading to tumor recurrence. The purpose of this review is to summarize the role of PCD and inflammatory TME in the tumor evolution of GBM and promising methods to overcome tumor evolution.
Article
Full-text available
Pharmacologically active salicylanilides (2-hydroxy-N-phenylbenzamides) have been a promising area of interest in medicinal chemistry-related research for quite some time. This group of compounds has shown a wide spectrum of biological activities, including but not limited to anticancer effects. In this study, substituted salicylanilides were chosen to evaluate the in vitro activity on U87 human glioblastoma (GBM) cells. The parent salicylanilide, salicylanilide 5-chloropyrazinoates, a 4-aminosalicylic acid derivative, and the new salicylanilide 4-formylbenzoates were chemically and in vitro characterized. To enhance the internalization of the compounds, they were conjugated to delivery peptides with the formation of oxime bonds. Oligotuftsins ([TKPKG]n, n = 1–4), the ligands of neuropilin receptors, were used as GBM-targeting carrier peptides. The in vitro cellular uptake, intracellular localization, and penetration ability on tissue-mimicking models of the fluorescent peptide derivatives were determined. The compounds and their peptide conjugates significantly decreased the viability of U87 glioma cells. Salicylanilide compound-induced GBM cell death was associated with activation of autophagy, as characterized by immunodetection of autophagy-related processing of light chain 3 protein.
Article
Aim: We predicted the modulation of autophagy and apoptosis in response to temozolomide (TMZ) and IFN-γ based on changes in the expression of non-coding RNAs in C6-induced glioblastoma (GBM). Materials & methods: Each rat received an intraperitoneal injection of TMZ (7.5 mg/kg) and/or IFN-γ (50,000 IU). Results: The reduced expression of H19 and colorectal neoplasia differentially expressed (CRNDE) was associated with a reduction in autophagy in response to TMZ, IFN-γ and TMZ + IFN-γ therapy, whereas the decreased level of miR-29a (proapoptotic miRNA) was associated with an increase in apoptosis. Conclusion: It appears that H19 promotes switching from autophagy to apoptosis in response to combination therapy of TMZ and IFN-γ through the miR-29a/autophagy-related protein 9A (ATG9A) pathway in C6-induced GBM.
Article
Glioma is the most common primary malignant brain tumour, and survival is poor. Hirudin has anticancer pharmacological effects through suppression of glioma cell progression, but the molecular target and mechanism are poorly understood. In this study, we observed that hirudin dose- and time-dependently inhibited glioma invasion, migration and proliferation. Mechanistically, hirudin activated LC3-II but not Caspase-3 to induce the autophagic death of glioma cells by decreasing the phosphorylation of mTOR and its downstream substrates ULK1, P70S6K and 4EBP1. Furthermore, hirudin inhibited glioma growth and induced changes in autophagy in cell-derived xenograft (CDX) nude mice, with a decrease in mTOR activity and activation of LC3-II. Collectively, our results highlight a new anticancer mechanism of hirudin in which hirudin-induced inhibition of glioma progression through autophagy activation is likely achieved by inhibition of the mTOR signalling pathway, thus providing a molecular basis for hirudin as a potential and effective clinical drug for glioma therapy.
Article
Full-text available
Background Host malignant stromal cells induced by glioma stem/progenitor cells were revealed to be more radiation-resistant than the glioma stem/progenitor cells themselves after malignant transformation in nude mice. However, the mechanism underlying this phenomenon remains unclear. Methods Malignant stromal cells induced by glioma stem/progenitor cell 2 (GSC-induced host brain tumor cells, ihBTC2) were isolated and identified from the double color-coded orthotopic glioma nude mouse model. The survival fraction at 2 Gy (SF2) was used to evaluate the radiation resistance of ihBTC2, the human glioma stem/progenitor cell line SU3 and its radiation-resistant sub-strain SU3-5R and the rat C6 glioma cell line. The mRNA of Notch 1 and Hes1 from ihBTC2 cells were detected using qPCR before and after 4 Gy radiation. The expression of the Notch 1, pAkt and Bcl-2 proteins were investigated by Western blot. To confirm the role of the Notch pathway in the radiation resistance of ihBTC2, Notch signaling blocker gamma secretase inhibitors (GSIs) were used. Results The ihBTC2 cells had malignant phenotypes, such as infinite proliferation, hyperpentaploid karyotype, tumorigenesis in nude mice and expression of protein markers of oligodendroglia cells. The SF2 of ihBTC2 cells was significantly higher than that of any other cell line (P<0.05, n = 3). The expression of Notch 1 and Hes1 mRNAs from ihBTC2 cells was significantly increased after radiation. Moreover, the Notch 1, pAkt and Bcl-2 proteins were significantly increased after radiation (P<0.05, n = 3). Inhibition of Notch signaling markedly enhanced the radiosensitivity of ihBTC2 cells. Conclusions In an orthotopic glioma model, the malignant transformation of host stromal cells was induced by glioma stem/progenitor cells. IhBTC2 cells are more radiation-resistant than the glioma stem/progenitor cells, which may be mediated by activation of the Notch signaling pathway.
Article
Full-text available
LC3s (MAP1-LC3A, B and C) are structural proteins of autophagosomal membranes, widely used as biomarkers of autophagy. Whether these three LC3 proteins have a similar biological role in autophagy remains obscure. We examine in parallel the subcellular expression patterns of the three LC3 proteins in a panel of human cancer cell lines, as well as in normal MRC5 fibroblasts and HUVEC, using confocal microscopy and western blot analysis of cell fractions. In the cytoplasm, there was a minimal co-localization between LC3A, B and C staining, suggesting that the relevant autophagosomes are formed by only one out of the three LC3 proteins. LC3A showed a perinuclear and nuclear localization, while LC3B was equally distributed throughout the cytoplasm and localized in the nucleolar regions. LC3C was located in the cytoplasm and strongly in the nuclei (excluding nucleoli), where it extensively co-localized with the LC3A and the Beclin-1 autophagy initiating protein. Beclin 1 is known to contain a nuclear trafficking signal. Blocking nuclear export function by Leptomycin B resulted in nuclear accumulation of all LC3 and Beclin-1 proteins, while Ivermectin that blocks nuclear import showed reduction of accumulation, but not in all cell lines. Since endogenous LC3 proteins are used as major markers of autophagy in clinical studies and cell lines, it is essential to check the specificity of the antibodies used, as the kinetics of these molecules are not identical and may have distinct biological roles. The distinct subcellular expression patterns of LC3s provide a basis for further studies.
Article
Macroautophagy is a dynamic process involving the rearrangement of subcellular membranes to sequester cytoplasm and organelles for delivery to the lysosome or vacuole where the sequestered cargo is degraded and recycled. This process takes place in all eukaryotic cells. It is highly regulated through the action of various kinases, phosphatases, and guanosine triphosphatases (GTPases). The core protein machinery that is necessary to drive formation and consumption of intermediates in the macroautophagy pathway includes a ubiquitin-like protein conjugation system and a protein complex that directs membrane docking and fusion at the lysosome or vacuole. Macroautophagy plays an important role in developmental processes, human disease, and cellular response to nutrient deprivation.
Article
Autophagy is a pathway mediating vacuolar degradation and recycling of proteins and organelles, which plays crucial roles in cellular physiology. To ensure its proper cytoprotective function, the induction and amplitude of autophagy are tightly regulated, and defects in its regulation are associated with various diseases. Transcriptional control of autophagy is a critical aspect of autophagy regulation, which remains largely unexplored. In particular, very few transcription factors involved in the activation or repression of autophagy-related gene expression have been characterized. To identify such regulators, we analyzed the expression of representative ATG genes in a large collection of DNA-binding mutant deletion strains in growing conditions as well as after nitrogen or glucose starvation. This analysis identified several proteins involved in the transcriptional control of ATG genes. Further analyses showed a correlation between variations in expression and autophagy magnitude, thus identifying new positive and negative regulators of the autophagy pathway. By providing a detailed analysis of the regulatory network of the ATG genes our study paves the way for future research on autophagy regulation and signaling.
Article
p62/SQSTM1 is a stress-inducible cellular protein that is conserved among metazoans but not in plants and fungi. p62/SQSTM1 has multiple domains that mediate its interactions with various binding partners, and it serves as a signaling hub for diverse cellular events such as amino-acid sensing and the oxidative stress response. In addition, p62/SQSTM1 functions as a selective autophagy receptor for degradation of ubiqutinated substrates. Herein we delineate the current knowledge about p62 with regard to mTORC1 activation, the Keap1-Nrf2 pathway, and selective autophagy. This article is protected by copyright. All rights reserved.
Article
Protein kinase C (PKC) is a family of serine/threonine kinases that regulate diverse cellular functions including cell death, proliferation and survival. Recent studies have reported that PKCdelta, are involved in apoptosis or autophagy induction. In the present study we focused on how PKCdelta regulates proliferation and cancer stem cell (CSC) properties of the hormone-independent mammary cancer cell line LM38-LP, using pharmacological and genetic approaches. We found that pharmacological inhibition of PKCdelta, by Rottlerin treatment, impairs in vitro LM38-LP proliferation through cell cycle arrest, inducing the formation of cytoplasmic-vacuoles. Using immunofluorescence we confirmed that Rottlerin treatment induced the apparition of LC3 dots in cell cytoplasm, and increased autophagy flux. On the other side, the same treatment increased CSC growth rate and self-renewal. Furthermore, Rottlerin pre-treatment induced in CSC the development of a "grape-like" morphology when they are growing in 3D cultures (Matrigel), usually associated with a malignant phenotype, as well as an increase in the number of experimental lung metastasis when these cells were inoculated in vivo. The PKCdelta knockdown, by RNA interference, induced autophagy and increased CSC number, indicating that these effects are indeed exerted through a PKCdelta dependent pathway. Finally, the increase in the number of mammospheres could be reversed by a 3MA treatment, suggesting that autophagy mechanism is necessary for the increased of CSC self-renewal induced by PKCdelta inhibition. Here we demonstrated that PKCdelta activity exerts a dual role through the autophagy mechanism, decreasing proliferative capacity of mammary tumor cells but also regulating tumor stem cell self-renewal. This article is protected by copyright. All rights reserved.
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
DNA double-strand breaks are the critical lesions responsible for the majority of ionizing radiation-induced cell killing. Thus, the ability of tumor cells to elicit a DNA damage response following radiation, via activation of DNA repair and cell-cycle checkpoints, promotes radiation resistance and tumor cell survival. Consequently, agents that target these DNA damage response pathways are being developed to overcome radiation resistance. Overall, these agents are effective radiosensitizers; however, their mechanisms of tumor cell selectivity are not fully elucidated. In this review, we focus on the crucial radiation-induced DNA damage responses as well as clinical and translational advances with agents designed to inhibit these responses. Importantly, we describe how synthetic lethality can provide tumor cell-selective radiosensitization by these agents and expand the therapeutic window for DNA damage response-targeted agents used in combination with radiotherapy. Clin Cancer Res; 21(13); 2898-904. ©2015 AACR. ©2015 American Association for Cancer Research.