ArticlePDF AvailableLiterature Review

Abstract and Figures

Simple Summary Glioblastoma (GB) is the deadliest type of primary brain tumor. Following diagnosis the patient´s median survival is only 16 months. There are currently around 450 clinical trials focused on the development of more effective therapies for GB. Nevertheless, radiotherapy remains the most clinically relevant and effective treatment for this devastating disease. Unfortunately, radiotherapy resistance (radioresistance) is frequently observed in GB patients. As a consequence tumor regrowth (recurrence) occurs and eventually the patient succumbs to the disease. It is crucial to fully understand the mechanisms by which GB cells become resistant to radiation in order to improve the sensitivity of these cells to radiotherapy and develop novel strategies to overcome this issue. In this review, we examined how low tumor oxygenation (known as hypoxia) which is a main feature of GB contributes to radioresistance to better understand the implications of this tumor microenvironment in GB treatment and recurrence. Abstract Glioblastoma (GB) (grade IV astrocytoma) is the most malignant type of primary brain tumor with a 16 months median survival time following diagnosis. Despite increasing attention regarding the development of targeted therapies for GB that resulted in around 450 clinical trials currently undergoing, radiotherapy still remains the most clinically effective treatment for these patients. Nevertheless, radiotherapy resistance (radioresistance) is commonly observed in GB patients leading to tumor recurrence and eventually patient death. It is therefore essential to unravel the molecular mechanisms underpinning GB cell radioresistance in order to develop novel strategies and combinational therapies focused on enhancing tumor cell sensitivity to radiotherapy. In this review, we present a comprehensive examination of the current literature regarding the role of hypoxia (O2 partial pressure less than 10 mmHg), a main GB microenvironmental factor, in radioresistance with the ultimate goal of identifying potential molecular markers and therapeutic targets to overcome this issue in the future.
This content is subject to copyright.
cancers
Review
The Role of Hypoxia in Glioblastoma Radiotherapy Resistance
Agathe L. Chédeville 1,2,3 and Patricia A. Madureira 4, *


Citation: Chédeville, A.L.;
Madureira, P.A. The Role of Hypoxia
in Glioblastoma Radiotherapy
Resistance. Cancers 2021,13, 542.
https://doi.org/10.3390/
cancers13030542
Academic Editor: David Wong
Received: 28 December 2020
Accepted: 29 January 2021
Published: 1 February 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1INSERM, UMR 1287, Gustave Roussy, CEDEX 94805 Villejuif, France;
Agathe.CHEDEVILLE@gustaveroussy.fr
2UniversitéParis-Saclay, UMR 1287, Gustave Roussy, CEDEX 94805 Villejuif, France
3Gustave Roussy, UMR 1287, 114, Rue Edouard-Vaillant, CEDEX 94805 Villejuif, France
4Centre for Biomedical Research (CBMR), University of Algarve, Gambelas Campus, Building 8, Room 2.22,
9005-139 Faro, Portugal
*Correspondence: patricia.madureira75@gmail.com or pamadureira@ualg.pt
Simple Summary:
Glioblastoma (GB) is the deadliest type of primary brain tumor. Following diag-
nosis the patient
´
s median survival is only 16 months. There are currently around 450 clinical trials
focused on the development of more effective therapies for GB. Nevertheless, radiotherapy remains
the most clinically relevant and effective treatment for this devastating disease. Unfortunately, radio-
therapy resistance (radioresistance) is frequently observed in GB patients. As a consequence tumor
regrowth (recurrence) occurs and eventually the patient succumbs to the disease. It is crucial to fully
understand the mechanisms by which GB cells become resistant to radiation in order to improve the
sensitivity of these cells to radiotherapy and develop novel strategies to overcome this issue. In this
review, we examined how low tumor oxygenation (known as hypoxia) which is a main feature of GB
contributes to radioresistance to better understand the implications of this tumor microenvironment
in GB treatment and recurrence.
Abstract:
Glioblastoma (GB) (grade IV astrocytoma) is the most malignant type of primary brain
tumor with a 16 months median survival time following diagnosis. Despite increasing attention
regarding the development of targeted therapies for GB that resulted in around 450 clinical trials
currently undergoing, radiotherapy still remains the most clinically effective treatment for these
patients. Nevertheless, radiotherapy resistance (radioresistance) is commonly observed in GB patients
leading to tumor recurrence and eventually patient death. It is therefore essential to unravel the
molecular mechanisms underpinning GB cell radioresistance in order to develop novel strategies and
combinational therapies focused on enhancing tumor cell sensitivity to radiotherapy. In this review,
we present a comprehensive examination of the current literature regarding the role of hypoxia (O
2
partial pressure less than 10 mmHg), a main GB microenvironmental factor, in radioresistance with
the ultimate goal of identifying potential molecular markers and therapeutic targets to overcome this
issue in the future.
Keywords:
glioblastoma (GB); hypoxia; radiotherapy; Hypoxia Inducible Factor (HIF); radioresis-
tance; glioma stem cells (GSC)
1. Introduction
Glioblastoma (GB) is classified by the World Health Organization (WHO) as a grade
IV astrocytoma. It is the deadliest primary malignant brain tumor; the median patient
survival time being only 16 months [13].
The current classification of the Central Nervous System (CNS) tumors by the WHO
combines both their histopathological name followed by the characteristic genetic signa-
ture [
4
]. In accordance to this, GB is classified as GB, IDH-wildtype which is the most
prevalent type, corresponding to approximately 90% of all cases, and GB, IDH-mutant.
Over 90% of GB develop de novo, called primary GB. However, a minority of GB develop
Cancers 2021,13, 542. https://doi.org/10.3390/cancers13030542 https://www.mdpi.com/journal/cancers
Cancers 2021,13, 542 2 of 16
slowly from low-grade astrocytomas (secondary GB). Mutations in IDH are more frequently
observed in secondary GB. There are three Isocitrate Dehydrogenase (IDH) enzymes (IDH1,
IDH2 and IDH3), but only IDH1 and IDH2 enzymes have been shown to be mutated in GB.
IDHs are responsible for the conversion of isocitrate to
α
-ketoglutarate. This process results
in the production of the reducing agent Nicotinamide Adenine Dinucleotide Phosphate
(NADPH). IDH-mutant enzymes have approximately 50% less activity compared to IDH
wild-type (WT) proteins. This results in impaired production of bioenergy (NADPH) and
intermediates and the production of the onco-metabolite 2-hydroxyglutarate (2-HG) which
causes epigenetic changes. 2-HG via hyper-methylation leads to the loss of differentiation
of GB cells. These changes caused by IDH mutation lead to reduced GB, IDH-mutant
tumor growth compared to GB, IDH-WT. This consequently translates to a better overall
prognosis for patients with GB, IDH-mutant [5].
GB has been further sub-classified into classical, proneural, neural and mesenchymal
sub-types based on specific genetic signatures [
6
,
7
]. In this way, the classical sub-type is
characterized by Epidermal Growth Factor Receptor (EGFR) gene amplification or mutation
(leading to a constitutively active receptor) as well as by over-expression of neural stem
cell genes. These include Sonic hedgehog,Notch and NES [
7
]. The proneural sub-type shows
a gene signature characterized by over-expression of many proneural genes (e.g., DCX,
SOX, TCF4, ASCL1 and DLL3), amplification of the Platelet-Derived Growth Factor Receptor
A(PDGFRA) gene and inactivation or loss of TP53 [
7
,
8
]. In addition, IDH mutations are
more common within the proneural subtype [
9
]. The expression of neuron gene markers,
such as NEFL,GABRA1,SYT1 and SLC12A5 is a hallmark of the neural sub-type [
7
]. Lastly,
the mesenchymal sub-type signature includes the expression of mesenchymal genes (e.g.,
CHI3L1 and MET) [
8
] as well as inactivating mutations or deletion of the Neurofibromin 1
(NF1) gene [7,10,11].
The current standard treatment for GB was established in 2005 by Roger Stupp and
colleagues [
12
]. The so called “Stupp protocol” encompasses GB resection surgery (when
possible as evaluated by MRI imaging) after what concurrent radiotherapy and chemother-
apy with temozolomide (TMZ) are implemented. This is followed by additional 6 cycles of
TMZ administration [
12
]. Radiotherapy alone can considerably increase patient survival.
However, beneficial effects of chemotherapy with TMZ are most commonly observed in a
sub-set of patients whose tumors contain O6-methylguanine DNA methyltransferase (MGMT)
promoter methylation [
1
,
9
]. MGMT promoter methylation is also a prognostic factor associ-
ated with longer survival irrespective of TMZ treatment as well as longer post-progression
survival (3–4 months) in patients with recurrent GB [
13
]. MGMT is a DNA repair enzyme
that fixes damaged guanine nucleotides (O6-methylguanine) via transferring the methyl
group at the O6 site of guanine to its cysteine residues. This reverts the gene mutation and
subsequently avoids cell death induced by alkylating agents such as TMZ [
14
]. Several
studies have demonstrated that regulation of MGMT expression in GB occurs mainly via
epigenetic modification, namely through the methylation of CpG islands within the MGMT
promoter. This leads to heterochromatinization which is accompanied by rearrangement
and random localization of nucleosomes. Consequently, binding of transcription factors to
the MGMT promoter becomes impaired [14].
The current standard radiotherapy dosage is a total of 60 Grays (Gy) in fractions of
2 Gy, administered 5 days a week for 6 weeks [
12
]. TMZ is concurrently administrated at a
dose of 75 mg/m
2
daily for 6 weeks. After a rest period of one month, TMZ chemotherapy
is restarted at a dose of 150 mg/m
2
daily for 5 days in the first month cycle. If this dose is
tolerated, it can be increased up to 200 mg/m
2
for 5 days per month. TMZ is administrated
for 6 months after radiotherapy, but many physicians continue TMZ administration for
12–18 months even though it has not been proved to increase overall survival [
12
,
15
].
Worryingly, almost all GB patients develop resistance to current therapy and eventually
succumb to the disease.
Despite the development of a large number of studies and hundreds of ongoing
clinical trials, GB treatment has not changed since 2005 [
9
,
12
]. Understanding the complex
Cancers 2021,13, 542 3 of 16
biology of GB and the role of the tumor microenvironment is therefore crucial to develop
novel and effective treatments in GB.
GB pathological features include hypoxic foci (where O
2
partial pressure is less than
10 mmHg) containing necrotic cores. The hypoxic areas are surrounded by cell pseudopal-
isades and microvascular hyperplasia [
9
]. Research suggests that cellular pseudopalisades
constitute invasive fronts of the tumor that likely originated from GB cells migrating away
from the hypoxic regions. These cells over-secrete proangiogenic factors leading to an
intensified form of angiogenesis which is known as microvascular hyperplasia [9].
The GB hypoxic microenvironment has been shown to be highly associated with
tumor invasion and resistance to chemo- and radiotherapy which are the main causes of
death in GB patients [
16
,
17
]. Importantly, dynamic contrast enhanced MRI analyses have
indicated that Hypoxia Inducible Factor 1 (HIF-1) (hypoxia marker) and Vascular Endothe-
lial Growth Factor A (VEGFA) staining and tumor vascularity significantly correlate with
worse progression-free and overall GB patient survival [18,19].
In this review article, we examined the existing literature regarding the role of hy-
poxia in supporting radiotherapy resistance in GB with the aim to better understand the
implications of this tumor microenvironment in GB treatment and recurrence.
2. Basic Principles of Cancer Radiotherapy
Radiotherapy is currently the major and most effective treatment modality for GB
patients. Nevertheless, radioresistance remains a major clinical problem for these patients.
Ionizing radiation (IR) was discovered just before the turn of the 20th century by
Marie and Pierre Curie and Wilhelm Conrad Roentgen [
20
]. It was during the 1920s that
cancer radiotherapy was piloted and significantly evolved [
20
] due to several technologic
and research advances. These included the invention by Coolidge et al. of a sealed-off
vacuum x-ray tube which could be operated at 180,000 to 200,000 volts which introduced
the kilovoltage era in radiotherapy. Another important advancement that contributed to
the effectiveness of radiotherapy at that time was the development of the first quantitative
methods for the measurement of radiation dose and of the first physical unit of dose, the
roentgen (later replaced by the rad). Lastly, the experimental and clinical radiobiology re-
search work led by Claude Regaud at the Fondation Curie in Paris pioneered the procedure
and development of fractionated radiotherapy which is still currently in use [20].
The ability of IR to kill tumor cells relies mainly on its DNA damaging effects. This
damage can occur either directly on the DNA molecules (accounting for 30–40% of lesions),
or indirectly through the generation of free radicals such as reactive oxygen species (ROS)
or reactive nitrogen species (RNS) that in turn damage the DNA molecules (responsible for
60–70% of lesions) [21,22].
Water radiolysis is a main process in the formation of free radicals (ROS) by IR,
resulting in the formation of electrons, atoms, OH˙ radicals, H
3
O
+
, OH
ions and
dihydrogen (H2) and hydrogen peroxide (H2O2) molecules [23].
IR produces a spectrum of DNA base lesions, the most prevalent being 8-oxo-guanaine
(8-oxoG), thymine glycol (5,6-dihydroxy-5,6-dihydrothymine) and formamidopyrimidines [4,6-
diamino-5-formamidopyrimidine (FapyAde) and 2,6-diamino-4-hydroxy-5-formamidopyrimidine
(FapyGua)] [
24
]. In addition, IR produces DNA Single Strand Breaks (SSBs) that have
a unique signature, generating 3
0
phosphate or 3
0
-phosphoglycolate ends rather than
3
0
-OH ends. Particularly important IR induced lesions are double strand breaks (DSBs)
which occur due to multiple damaged sites closely located on both strands of the DNA
molecule [
24
]. DSBs are more difficult to repair compared to SSBs leading to cancer cell
death [
25
]. DSBs typically trigger DNA-damage responses (DDR). However, when DSBs
cannot be efficiently repaired by the cellular DDR mechanisms, irradiated cells undergo the
so-called mitotic catastrophe which is a major cell death mechanism caused by IR-induced
DNA damage [26].
Typically, a dose of 1Gy of X-ray radiation produces around 3000 damaged bases,
1000 SSBs and 40 DSBs [22].
Cancers 2021,13, 542 4 of 16
The decreased oxygen levels observed in hypoxic GB cells, lead to resistance to IR due
to the reduced availability of oxygen which is needed to stabilize the DNA strand breaks
caused by radiotherapy [
27
]. Under normoxic conditions (physiological or normal oxygen
levels, where tissue oxygenation is around 40 mmHg), cells are vulnerable to IR due to
oxygen fixation leading to irreversible DNA damage. However, under hypoxic conditions
(where O
2
partial pressure (pO
2
) is below 10 mmHg), there is diminished production
of DNA radicals due to the low levels of oxygen and subsequently cells become more
resistant to radiotherapy [
28
]. Consequently, the radiation dose required to achieve the
same biological effect is about three times higher in the absence of oxygen as compared to
physiological oxygen levels [27].
3. Hypoxia in GB
Tumor hypoxia is a hallmark of GB and mainly occurs due to the abnormal neo-
vascularization observed within these tumors [
29
]. The blood vessels that feed the GB
are typically highly permeable and easily collapsible due to the excessive recruitment
and proliferation of endothelial cells (caused by excessive secretion of VEGFA by the
tumor cells) and lack of pericyte coverage (which are cells that provide support to the
blood vessels) [
30
32
]. In addition, these vessels exhibit larger diameters and possess
thicker basement membranes when compared to physiological brain blood vessels [
33
].
As a consequence, the occurrence of microvascular thromboses and vessel occlusions are
frequently observed in GB [
34
] which significantly impede blood flow leading to a het-
erogeneous microenvironment regarding tumor oxygenation [
35
]. Moreover, the anarchic
organization and instability of the vascular system within the tumor can lead to dynamic
phases of hypoxia and then reoxygenation within the different tumor fractions, known as
“cycling hypoxia” [36].
3.1. Regulation of HIF Transcription Factors
The cellular response to hypoxic stress is largely orchestrated by the HIF transcription
factors. HIFs are heterodimers composed of an
α
subunit (e.g., HIF-1
α
, EPAS1/HIF-2
α
,
or HIF-3
α
) which is negatively regulated by oxygen (O
2
) and a
β
subunit, HIF-1
β
, also
known as aryl hydrocarbon receptor nuclear translocator (ARNT), which is expressed
constitutively in cells [
37
,
38
]. Regarding protein domain structures, HIF-1
α
, HIF-2
α
, and
HIF-1
β
subunits all have a Per-Arnt-Sim (PAS) and a basic Helix-Loop-Helix (bHLH) do-
mains which are involved in the heterodimer assembly and binding to Hypoxia Responsive
Elements (HRE) within HIF target gene promoters and a characteristic C-Terminal Domain
(C-TAD) (Figure 1). Each HIF-
α
subunit also contain an Oxygen-Dependent Degradation
Domain (ODDD) that is involved in the degradation of these proteins in the presence of
normal levels of oxygen, and a specific N-Terminal Domain (N-TAD) (Figure 1). N-TAD
and C-TAD domains are able to interact with p300/CBP HIF transcriptional coactiva-
tors [
39
41
]. Different variants of the HIF-3
α
subunit containing diverse deletions of the
domains described above have been shown to exist (reviewed in [42]).
HIF transcriptional activity is highly dependent on the degradation (negative regu-
lation) or stabilization (positive regulation) of the HIF-
α
subunit which is regulated by
intracellular levels of oxygen. In normoxic cells, the Prolyl Hydroxylases 1-3 (PHD1-3)
hydroxylate two prolyl residues within the HIF-
α
subunit. This enables the binding of the
Von Hippel-Lindau (VHL) protein to the HIF-
α
subunit and the subsequent recruitment of
E3 ubiquitin ligases that target HIF-
α
for degradation via the proteasome (
Figure 2
) [
43
45
].
Hypoxia inhibits PHD activity and consequently leads to HIF-
α
stabilization and transloca-
tion to the nucleus where it binds to the HIF-1
β
subunit and p300/CBP cofactors. HIFs bind
to HRE within their target gene promoters orchestrating the hypoxic response [
46
48
]. The
hydroxylase, Factor-Inhibiting HIF (FIH) has also been shown to regulate HIF activity in
an oxygen dependent manner. In normoxic cells, FIH hydroxylates an asparagine residue
within the HIF-
α
subunit which impairs the interaction between HIF and the transcrip-
Cancers 2021,13, 542 5 of 16
tional activators, p300/CBP subsequently negatively impacting on HIF transcriptional
activity (Figure 2) [49,50].
Cancers 2021, 13, x 4 of 16
The decreased oxygen levels observed in hypoxic GB cells, lead to resistance to IR
due to the reduced availability of oxygen which is needed to stabilize the DNA strand
breaks caused by radiotherapy [27]. Under normoxic conditions (physiological or normal
oxygen levels, where tissue oxygenation is around 40 mmHg), cells are vulnerable to IR
due to oxygen fixation leading to irreversible DNA damage. However, under hypoxic
conditions (where O2 partial pressure (pO2) is below 10 mmHg), there is diminished pro-
duction of DNA radicals due to the low levels of oxygen and subsequently cells become
more resistant to radiotherapy [28]. Consequently, the radiation dose required to achieve
the same biological effect is about three times higher in the absence of oxygen as compared
to physiological oxygen levels [27].
3. Hypoxia in GB
Tumor hypoxia is a hallmark of GB and mainly occurs due to the abnormal neovas-
cularization observed within these tumors [29]. The blood vessels that feed the GB are
typically highly permeable and easily collapsible due to the excessive recruitment and
proliferation of endothelial cells (caused by excessive secretion of VEGFA by the tumor
cells) and lack of pericyte coverage (which are cells that provide support to the blood ves-
sels) [30–32]. In addition, these vessels exhibit larger diameters and possess thicker base-
ment membranes when compared to physiological brain blood vessels [33]. As a conse-
quence, the occurrence of microvascular thromboses and vessel occlusions are frequently
observed in GB [34] which significantly impede blood flow leading to a heterogeneous
microenvironment regarding tumor oxygenation [35]. Moreover, the anarchic organiza-
tion and instability of the vascular system within the tumor can lead to dynamic phases
of hypoxia and then reoxygenation within the different tumor fractions, known as “cy-
cling hypoxia [36].
3.1. Regulation of HIF Transcription Factors
The cellular response to hypoxic stress is largely orchestrated by the HIF transcrip-
tion factors. HIFs are heterodimers composed of an α subunit (e.g., HIF-1α, EPAS1/HIF-
2α, or HIF-3α) which is negatively regulated by oxygen (O2) and a β subunit, HIF-1β, also
known as aryl hydrocarbon receptor nuclear translocator (ARNT), which is expressed
constitutively in cells [37,38]. Regarding protein domain structures, HIF-1α, HIF-2α, and
HIF-1β subunits all have a Per-Arnt-Sim (PAS) and a basic Helix-Loop-Helix (bHLH) do-
mains which are involved in the heterodimer assembly and binding to Hypoxia Respon-
sive Elements (HRE) within HIF target gene promoters and a characteristic C-Terminal
Domain (C-TAD) (Figure 1). Each HIF-α subunit also contain an Oxygen-Dependent Deg-
radation Domain (ODDD) that is involved in the degradation of these proteins in the pres-
ence of normal levels of oxygen, and a specific N-Terminal Domain (N-TAD) (Figure 1).
N-TAD and C-TAD domains are able to interact with p300/CBP HIF transcriptional coac-
tivators [39–41]. Different variants of the HIF-3α subunit containing diverse deletions of
the domains described above have been shown to exist (reviewed in [42]).
Figure 1. HIF protein domain structures. HIF-1α, HIF-2α, and HIF-1β subunits contain a bHLH domain (blue box), a PAS
domain (orange box) and a C-TAD domain (yellow box). In addition, HIF-1α and HIF-2α subunits contain an ODDD (red
box) and N-TAD (yellow box) domains. Different variants of the HIF-3α subunit containing diverse deletions of the do-
mains shown in the figure have been shown to exist.
Figure 1.
HIF protein domain structures. HIF-1
α
, HIF-2
α
, and HIF-1
β
subunits contain a bHLH domain (blue box), a
PAS domain (orange box) and a C-TAD domain (yellow box). In addition, HIF-1
α
and HIF-2
α
subunits contain an ODDD
(red box) and N-TAD (yellow box) domains. Different variants of the HIF-3
α
subunit containing diverse deletions of the
domains shown in the figure have been shown to exist.
Cancers 2021, 13, x 5 of 16
HIF transcriptional activity is highly dependent on the degradation (negative regu-
lation) or stabilization (positive regulation) of the HIF-α subunit which is regulated by
intracellular levels of oxygen. In normoxic cells, the Prolyl Hydroxylases 1-3 (PHD1-3)
hydroxylate two prolyl residues within the HIF-α subunit. This enables the binding of the
Von Hippel-Lindau (VHL) protein to the HIF-α subunit and the subsequent recruitment
of E3 ubiquitin ligases that target HIF-α for degradation via the proteasome (Figure 2)
[4345]. Hypoxia inhibits PHD activity and consequently leads to HIF-α stabilization and
translocation to the nucleus where it binds to the HIF-1β subunit and p300/CBP cofactors.
HIFs bind to HRE within their target gene promoters orchestrating the hypoxic response
[46–48]. The hydroxylase, Factor-Inhibiting HIF (FIH) has also been shown to regulate HIF
activity in an oxygen dependent manner. In normoxic cells, FIH hydroxylates an aspara-
gine residue within the HIF-α subunit which impairs the interaction between HIF and the
transcriptional activators, p300/CBP subsequently negatively impacting on HIF transcrip-
tional activity (Figure 2) [49,50].
Figure 2. Regulation of HIF transcription factors. In normoxic cells, the PHD1-3 hydroxylate two prolyl residues within
the HIF-α subunit enabling the binding of the VHL protein to the HIF-α subunit. VHL recruits E3 ubiquitin ligases that
target HIF-α for degradation via the proteasome. In hypoxic cells, PHD1-3 activity (which is oxygen dependent) is inhib-
ited. This leads to HIF-α stabilization and translocation to the nucleus where it binds to the HIF-1β subunit and p300/CBP
cofactors. HIFs bind to HRE within their target gene promoters. In normoxic cells, FIH hydroxylates an asparagine residue
within the HIF-α subunit. This blocks the interaction between HIF and the transcriptional activators, p300/CBP.
HIF-1 and HIF-2 are considered the main regulators of the hypoxia response [37,38],
while the existence of multiple variants of HIF-3α has highlighted that HIF-3 can function
in some cases as a transcriptional activator whereas certain variants can act as dominant
negative regulators of HIF-1 and/or HIF-2 transcriptional functions [42,51].
3.2. Hypoxia Independent HIF Activation in GB
Several genetic alterations leading to HIF activation, even in the absence of hypoxia,
have been reported in GB (Figure 3). These include the activation of the EGFR (either by
amplification or mutation of the EGFR gene) and the loss of the tumor suppressor genes
TP53 and Phosphatase and Tensin homolog (PTEN) [52–54]. The most common EGFR mu-
tation observed in GB is the deletion of exons 2–7 (EGFRvIII) resulting in the expression
of a constitutively active and ligand independent EGFRvIII receptor [55]. EGFR signaling
leads to the up-regulation of HIF-1α levels via the activation of the PI3K/AKT/mTOR
pathway [56,57]. Depletion of PTEN has been reported in about 20–40% of GB [53]. PTEN
constitutes the main negative regulator of the PI3K/AKT signaling pathway. Therefore,
loss of PTEN promotes the up-regulation of HIF-1α due to enhanced activity of the
PI3K/AKT/mTOR pathway which is observed in the absence of PTEN protein. Loss of the
Figure 2.
Regulation of HIF transcription factors. In normoxic cells, the PHD1-3 hydroxylate two prolyl residues within the
HIF-
α
subunit enabling the binding of the VHL protein to the HIF-
α
subunit. VHL recruits E3 ubiquitin ligases that target
HIF-
α
for degradation via the proteasome. In hypoxic cells, PHD1-3 activity (which is oxygen dependent) is inhibited. This
leads to HIF-
α
stabilization and translocation to the nucleus where it binds to the HIF-1
β
subunit and p300/CBP cofactors.
HIFs bind to HRE within their target gene promoters. In normoxic cells, FIH hydroxylates an asparagine residue within the
HIF-αsubunit. This blocks the interaction between HIF and the transcriptional activators, p300/CBP.
HIF-1 and HIF-2 are considered the main regulators of the hypoxia response [
37
,
38
],
while the existence of multiple variants of HIF-3
α
has highlighted that HIF-3 can function
in some cases as a transcriptional activator whereas certain variants can act as dominant
negative regulators of HIF-1 and/or HIF-2 transcriptional functions [42,51].
3.2. Hypoxia Independent HIF Activation in GB
Several genetic alterations leading to HIF activation, even in the absence of hypoxia,
have been reported in GB (Figure 3). These include the activation of the EGFR (either by am-
plification or mutation of the EGFR gene) and the loss of the tumor suppressor genes TP53
and Phosphatase and Tensin homolog (PTEN) [
52
54
]. The most common EGFR mutation
observed in GB is the deletion of exons 2–7 (EGFRvIII) resulting in the expression of a consti-
tutively active and ligand independent EGFRvIII receptor [
55
]. EGFR signaling leads to the
up-regulation of HIF-1
α
levels via the activation of the PI3K/AKT/mTOR pathway [
56
,
57
].
Depletion of PTEN has been reported in about 20–40% of GB [
53
]. PTEN constitutes the
main negative regulator of the PI3K/AKT signaling pathway. Therefore, loss of PTEN
Cancers 2021,13, 542 6 of 16
promotes the up-regulation of HIF-1
α
due to enhanced activity of the PI3K/AKT/mTOR
pathway which is observed in the absence of PTEN protein. Loss of the TP53 gene has
been linked to HIF-1
α
stabilization due to down-regulation of MDM2 transcription and
subsequent inhibition of MDM2 mediated ubiquitination and degradation of HIF-1
α
(Figure 3) [54].
Cancers 2021, 13, x 6 of 16
TP53 gene has been linked to HIF-1α stabilization due to down-regulation of MDM2 tran-
scription and subsequent inhibition of MDM2 mediated ubiquitination and degradation
of HIF-1α (Figure 3) [54].
Figure 3. Genetic alterations leading to HIF activation in GB. Activation of the Epidermal Growth
Factor Receptor (EGFR) by EGFR gene mutation (e.g., EGFRvIII) and/or amplification frequently
occurs in GB cells resulting in activation of the PI3K/AKT/mTOR pathway with the subsequent
accumulation of HIF-1α levels. The tumor suppressor Phosphatase and Tensin homolog (PTEN)
gene is deleted in 20–40% of GBs. PTEN is the main inhibitor of the PI3K/AKT signaling pathway.
Consequently, loss of PTEN will also lead to accumulation of HIF-1α via the PI3K/AKT/mTOR
pathway. Loss of p53 inhibits MDM2-mediated ubiquitination of HIF-1α leading to its accumula-
tion and increased HIF activity in GB cells.
3.3. HIF Transcriptional Targets in GB
HIFs induce the transcription of hundreds of genes involved in the regulation of main
cellular processes including angiogenesis, glycolysis, autophagy, motility and invasion,
chemo- and radioresistance [58,59].
Several studies using GB cell lines and/or clinical samples support a hypoxia trig-
gered metabolic switch towards glycolysis including the up-regulation of HK2, PFKFB3,
PFKFB4, PFKFP, LDHA, PDK1, SLC2A1/GLUT-1, CA9/CA IX, PGAM1, ENO1, ENO2,
ALDOA and SLC16A3/ MCT-4 genes and proteins (Figure 4) [9,59,60]. Hypoxic up-regu-
lation of many pro-angiogenic genes and proteins is also commonly observed in GB. These
include VEGFA, VEGFC, VEGFD, PGF/PlGF, ADM and ANGPTL4 (Figure 4)
[32,59,61,62]. GB is a highly invasive tumor and hypoxia has been shown to induce pro-
teins of the plasminogen system. These include the plasminogen receptor, S100A10, the
receptor for the urokinase Plasminogen Activator (uPA), uPAR and the Plasminogen Ac-
tivator Inhibitor-1 (PAI-1) (Figure 4) [59,63,64]. The co-localization of S100A10 with uPAR
Figure 3.
Genetic alterations leading to HIF activation in GB. Activation of the Epidermal Growth
Factor Receptor (EGFR) by EGFR gene mutation (e.g., EGFRvIII) and/or amplification frequently
occurs in GB cells resulting in activation of the PI3K/AKT/mTOR pathway with the subsequent
accumulation of HIF-1
α
levels. The tumor suppressor Phosphatase and Tensin homolog (PTEN)
gene is deleted in 20–40% of GBs. PTEN is the main inhibitor of the PI3K/AKT signaling pathway.
Consequently, loss of PTEN will also lead to accumulation of HIF-1
α
via the PI3K/AKT/mTOR
pathway. Loss of p53 inhibits MDM2-mediated ubiquitination of HIF-1
α
leading to its accumulation
and increased HIF activity in GB cells.
3.3. HIF Transcriptional Targets in GB
HIFs induce the transcription of hundreds of genes involved in the regulation of main
cellular processes including angiogenesis, glycolysis, autophagy, motility and invasion,
chemo- and radioresistance [58,59].
Several studies using GB cell lines and/or clinical samples support a hypoxia triggered
metabolic switch towards glycolysis including the up-regulation of HK2, PFKFB3, PFKFB4,
PFKFP, LDHA, PDK1, SLC2A1/GLUT-1, CA9/CA IX, PGAM1, ENO1, ENO2, ALDOA
and SLC16A3/ MCT-4 genes and proteins (Figure 4) [
9
,
59
,
60
]. Hypoxic up-regulation of
many pro-angiogenic genes and proteins is also commonly observed in GB. These include
VEGFA, VEGFC, VEGFD, PGF/PlGF, ADM and ANGPTL4 (Figure 4) [
32
,
59
,
61
,
62
]. GB is a
highly invasive tumor and hypoxia has been shown to induce proteins of the plasminogen
system. These include the plasminogen receptor, S100A10, the receptor for the urokinase
Cancers 2021,13, 542 7 of 16
Plasminogen Activator (uPA), uPAR and the Plasminogen Activator Inhibitor-1 (PAI-1)
(Figure 4) [
59
,
63
,
64
]. The co-localization of S100A10 with uPAR at the outer cell membrane
has been shown to promote the generation of the serine protease, plasmin by putting
plasminogen (inactive form of plasmin) and its activator, uPA into close proximity. This
leads to the subsequent degradation of the Extra-Cellular Matrix (ECM) by plasmin which
is a critical step in GB cell invasion. Importantly, plasmin also has the capacity to cleave
and activate many pro-MMPs, further accelerating ECM degradation [
65
,
66
]. The GB
hypoxic environment has also been shown to promote the up-regulation of autophagy
genes including BNIP-3 and DDIT4 (Figure 4) [
59
,
67
,
68
]. Several studies support that
during hypoxic stress autophagy allows the recycling of cellular components which is
critical for cell survival under oxygen and nutrients limiting conditions. Association
of GB hypoxia with chemoresistance has also been demonstrated. Several reports have
shown that hypoxic induction of ANGPTL4, DDIT4 and NDRG1 lead to resistance to
chemotherapy (Figure 4) [59,6973].
Cancers 2021, 13, x 7 of 16
at the outer cell membrane has been shown to promote the generation of the serine prote-
ase, plasmin by putting plasminogen (inactive form of plasmin) and its activator, uPA into
close proximity. This leads to the subsequent degradation of the Extra-Cellular Matrix
(ECM) by plasmin which is a critical step in GB cell invasion. Importantly, plasmin also
has the capacity to cleave and activate many pro-MMPs, further accelerating ECM degra-
dation [65,66]. The GB hypoxic environment has also been shown to promote the up-reg-
ulation of autophagy genes including BNIP-3 and DDIT4 (Figure 4) [59,67,68]. Several
studies support that during hypoxic stress autophagy allows the recycling of cellular com-
ponents which is critical for cell survival under oxygen and nutrients limiting conditions.
Association of GB hypoxia with chemoresistance has also been demonstrated. Several re-
ports have shown that hypoxic induction of ANGPTL4, DDIT4 and NDRG1 lead to re-
sistance to chemotherapy (Figure 4) [59,6973].
Figure 4. Hypoxia induced gene/protein expression in GB cells. Necrotic cells are represented in grey, hypoxic cells are
represented in purple, and normoxic cells are represented in peach color.
4. The Role of Hypoxia in GB Radioresistance
The molecular mechanisms by which GB becomes resistant to radiotherapy are still
not fully understood. However, it has been shown that radioresistance can at least in part
be due to the presence of hypoxic regions within the tumor [74].
Hypoxia contributes to radioresistance by controlling several cellular processes in-
cluding regulation of the cell cycle, inhibition of apoptosis and senescence, regulation of
autophagy and antioxidant/redox activity, promoting invasion and cancer cell stemness.
In addition, radiotherapy is more efficient in rapidly proliferating cells as compared to
slow-proliferating, quiescent and stem-like cells that are localized in the most hypoxic re-
gions of the tumor [28].
4.1. The Role of Cell Cycle Regulation Proteins in Hypoxia Induced Radioresistance
Several molecular mechanisms involved in cell cycle regulation have been shown to
play a role in hypoxia induced radioresistance in GB (Figures 5 and 6). A study showed
that MEK/ERK inhibition either by treatment with the drug, U0126 or downregulation of
ERK by siRNA significantly enhanced the radiosensitivity of hypoxic T98G, U87MG and
Figure 4.
Hypoxia induced gene/protein expression in GB cells. Necrotic cells are represented in grey, hypoxic cells are
represented in purple, and normoxic cells are represented in peach color.
4. The Role of Hypoxia in GB Radioresistance
The molecular mechanisms by which GB becomes resistant to radiotherapy are still
not fully understood. However, it has been shown that radioresistance can at least in part
be due to the presence of hypoxic regions within the tumor [74].
Hypoxia contributes to radioresistance by controlling several cellular processes in-
cluding regulation of the cell cycle, inhibition of apoptosis and senescence, regulation of
autophagy and antioxidant/redox activity, promoting invasion and cancer cell stemness.
In addition, radiotherapy is more efficient in rapidly proliferating cells as compared to
slow-proliferating, quiescent and stem-like cells that are localized in the most hypoxic
regions of the tumor [28].
4.1. The Role of Cell Cycle Regulation Proteins in Hypoxia Induced Radioresistance
Several molecular mechanisms involved in cell cycle regulation have been shown to
play a role in hypoxia induced radioresistance in GB (Figures 5and 6). A study showed
Cancers 2021,13, 542 8 of 16
that MEK/ERK inhibition either by treatment with the drug, U0126 or downregulation of
ERK by siRNA significantly enhanced the radiosensitivity of hypoxic T98G, U87MG and
U138MG GB cells [
75
]. Using a combination of siRNA approaches and chemical inhibitors
these authors further mapped the MEK/ERK/DNA-PKc/HIF-1
α
functional interplay in
hypoxia dependent GB radioresistance.
Cancers 2021, 13, x 8 of 16
U138MG GB cells [75]. Using a combination of siRNA approaches and chemical inhibitors
these authors further mapped the MEK/ERK/DNA-PKc/HIF-1α functional interplay in
hypoxia dependent GB radioresistance.
Figure 5. The role of hypoxia in GB radioresistance. Cell cycle regulation, glycolysis and Re-
dox/ROS regulatory mechanisms have been shown to support hypoxia dependent radioresistance
in GB cells.
Figure 6. Hypoxia induced mechanisms leading to radioresistance in GB. The hypoxic environment induces several mech-
anisms involved in radioresistance including mechanisms involved in cell cycle regulation, Redox regulation, glycolysis
and the maintenance of GSCs.
Figure 5.
The role of hypoxia in GB radioresistance. Cell cycle regulation, glycolysis and Redox/ROS regulatory mechanisms
have been shown to support hypoxia dependent radioresistance in GB cells.
Cancers 2021, 13, x 8 of 16
U138MG GB cells [75]. Using a combination of siRNA approaches and chemical inhibitors
these authors further mapped the MEK/ERK/DNA-PKc/HIF-1α functional interplay in
hypoxia dependent GB radioresistance.
Figure 5. The role of hypoxia in GB radioresistance. Cell cycle regulation, glycolysis and Re-
dox/ROS regulatory mechanisms have been shown to support hypoxia dependent radioresistance
in GB cells.
Figure 6. Hypoxia induced mechanisms leading to radioresistance in GB. The hypoxic environment induces several mech-
anisms involved in radioresistance including mechanisms involved in cell cycle regulation, Redox regulation, glycolysis
and the maintenance of GSCs.
Figure 6.
Hypoxia induced mechanisms leading to radioresistance in GB. The hypoxic environment induces several
mechanisms involved in radioresistance including mechanisms involved in cell cycle regulation, Redox regulation, glycolysis
and the maintenance of GSCs.
Cancers 2021,13, 542 9 of 16
Another report highlighted the role of the Fibroblast Growth Factor Receptor 1 (FGFR1)
in Phospholipase C Gamma (PlC
γ
)/HIF1
α
-dependent GB radioresistance [
76
]. FGFR
signalling is involved in the regulation of cell proliferation, differentiation, migration,
angiogenesis and tissue injury repair. PlC
γ
binds through its SH2 domain to a phos-
photyrosine residue within the C-terminal tail of FGFRs and is phosphorylated at the
tyrosine residues by the activated receptor tyrosine kinase. This phosphorylation activates
PlC
γ
which then hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) to generate
inositol1,4,5-trisphosphate (IP3) and 1,2-diacylglycerol (DAG) leading to Protein Kinase C
(PKC) activation. Using
in vitro
and
in vivo
knockdown approaches, the authors were able
to demonstrate that FGFR1/PlC
γ
/HIF1
α
signalling pathway confers radioresistance to
GB cells (e.g., U87, LN18) and derived tumor mouse xenografts via controlling radiation-
induced centrosome overduplication and radiation-induced mitotic cell death.
We recently observed a significant increase in DDIT4 expression in hypoxic GB cells
(e.g., U87, SEBTA-003, SEBTA-023, UP-007, UP-029) as compared to their normoxic coun-
terpart control cells [
59
]. These data supported a previous report using U87 cells [
67
]. We
also observed high expression of DDIT4 in GB patient clinical samples, particularly in GB
hypoxic core region as compared to normal brain specimens [
59
]. DDIT4 gene product
is the protein, REDD1 which is involved in the activation of the Tuberous Sclerosis 1/2
(TSC1/TSC2) complex, a main negative regulator of mTORC1 [
77
]. Even though over-
expression of REDD1 in GB has been linked to chemo- and radioresistance the molecular
mechanisms involved in these outcomes are still not fully understood [70,78].
Phosphorylation of 4E-BP1 occurs after activation of mTORC1, which functions down-
stream of the PI3K/AKT and AMPK kinase signaling pathways [
79
]. As previously men-
tioned, the PI3K/AKT signaling pathway is frequently activated in GB. Consequently, this
leads to increased rates of cap-dependent translation in an mTORC1/4E-BP1–dependent
manner. Using a mouse U87 GB xenograft model, a study has shown that 4E-BP1 promotes
hypoxia dependent radioresistance [
80
]. These authors found that loss of 4E-BP1 expres-
sion by siRNA did not significantly affect
in vitro
growth of U87 GB cells, but significantly
enhanced the growth of U87 tumor xenografts. Furthermore, 4E-BP1 knockdown U87
cells were significantly more sensitive to hypoxia-induced
in vitro
cell death. Most impor-
tantly, 4E-BP1 knockdown cells produced tumors with reduced fractions of radioresistant
hypoxic cells.
The exosomal secretion of micro-RNA-301-a (miR-301-a) by hypoxic GB cells (e.g.,
U87, LN229, U251) has also been shown to lead to radioresistance [
81
]. Importantly, clinical
analysis revealed higher levels of miR-301a expression in glioma samples with high HIF-1
α
levels and the percentage of serum exosomal miR-301a (low versus high) was distributed
according to the HIF-1
α
immunohistochemistry score.
In vitro
studies demonstrated that
miR-301a expression is regulated by HIF-1
α
. Furthermore, miR-301a directly repressed the
promoter of the tumor suppressor gene, TCEAL7 whose encoded protein binds to
β-catenin
inhibiting its translocation from the cytoplasm to the nucleus. Therefore, negatively
regulating the Wnt/
β
-catenin signaling pathway. In summary, targeting of TCEAL7 gene
expression by miR-301a secreted by hypoxic GB cells induced enhanced activation of the
Wnt/β-catenin signaling axis leading to radioresistance.
4.2. The Role of Glycolysis in Hypoxia Induced Radioresistance
As aforementioned, hypoxia induces a metabolic reprogramming towards glycolysis
in GB cells. Interestingly, a research study has shown that modulation of the glucose
metabolism can sensitize GB cells to IR (Figures 5and 6) [
82
]. Treatment of GB cells (e.g.,
U87, U251, LN229, DBTRG) with dichloroacetate, a PDK inhibitor, in combination with
radiotherapy reversed the radiotherapy-induced glycolytic shift in these cells and inhibited
their clonogenicity
in vitro
. Investigation into the molecular mechanism of action revealed
that dichloroacetate sensitized GB cells to radiotherapy by inducing G2–M phase cell-cycle
arrest, reducing mitochondrial reserve capacity, and increasing oxidative stress and DNA
damage in these cells [
82
].
In vivo
studies using a mouse xenograft model showed that
Cancers 2021,13, 542 10 of 16
radiotherapy in combination with dichloroacetate improved the survival of orthotopic GB-
bearing mice [
82
]. Taking into account that hypoxia constitutes a major microenvironmental
factor that triggers glycolysis in GB (including the specific up-regulation of PDK1 [
59
]) this
report provides encouraging data regarding targeting the glycolytic metabolism in order to
sensitize hypoxic GB cells to radiotherapy.
4.3. The Role of ROS Regulatory Systems in Hypoxia Induced Radioresistance
Several studies have shown that the regulation of intracellular ROS levels plays a
key role in hypoxic GB radioresistance (Figures 5and 6). ROS are radical and non-radical
oxygen-containing chemical molecules with different degrees of reactivity, including bio-
logically relevant molecules such as superoxide anion (O
2
), hydroxyl radical (
·
OH) and
hydrogen peroxide (H
2
O
2
) [
58
]. Of note, H
2
O
2
constitutes a key second messenger in many
cell signaling pathways [
58
,
83
]. However, due to their reactive properties ROS contribute
to protein oxidation, lipid peroxidation and/or DNA damage that can ultimately result
in either cell death or tumorigenesis (due to DNA mutagenesis) [
58
]. To overcome this
issue, cells possess several antioxidant systems that inactivate ROS and recycle oxidized
molecules (reviewed in [
58
]). A study showed that exposure of T98G GB cells to cycling
hypoxia induced the up-regulation of the aspartate-aminotransferase Glutamic-Oxaloacetic
Transaminase 1 (GOT1) protein, leading to increased levels of the antioxidant protein,
glutathione (GSH), decreased intracellular ROS levels and enhanced radioresistance [
84
].
Most importantly, targeting glutamine-dependent antioxidant capacity or glutathione
metabolism reversed the cycling hypoxia induced GB cells radioresistance. Exposure to
either acute or cycling hypoxia was shown to trigger the up-regulation of the mitochondrial
Citrate Carrier (CIC) and IDH2 in T98G GB cells
in vitro
[
85
]. CIC protein belongs to the
large family of mitochondrial metabolite carriers. It is a mitochondrial transmembrane
protein whose main function is to mediate the exchange of mitochondrial citrate for cy-
tosolic malate. This process is accompanied with the transport of one proton and therefore
can influence the mitochondrial membrane potential [
86
]. In fact, the tumorigenic activity
of CIC has been shown to be linked to its role in mitochondrial membrane integrity [
87
]
which is crucial to inhibit ROS induced apoptosis.
NADPH oxidase subunit 4 (Nox4) has been shown to mediate cycling (intermittent)
hypoxia induced radioresistance in GB cells [
88
]. In this report, the GBM8401 and U251
cell lines were stably transfected with a dual hypoxia HIF-1 signaling reporter construct.
The mouse tumor xenograft studies showed that Nox4 was highly expressed in the cycling
hypoxic areas within the tumor microenvironment. In addition, when compared to the
normoxic or acutely hypoxic GB cells, the cycling hypoxic GB cells derived from tumor
xenografts showed significantly higher expression of Nox4, enhanced ROS levels and
increased radioresistance which was reversed by Nox4 suppression in intracerebral GB
bearing mice [88].
Another report revealed that hypoxia increased U87 GB cell radioresistance
in vitro
and
in vivo
via long-term induction of HIF-1 signaling transduction in a ROS dependent-
manner [
89
]. These authors performed clonogenic survival assays to show that hypoxia
pretreatment of U87 cells significantly increased GB cell resistance to IR compared with
normoxic U87 control cells. To determine whether HIF-1 was a crucial mediator of hypoxia-
induced radioresistance in U87 cells, they used a HIF-1 siRNA approach. Hypoxic HIF-1
knockdown U87 cells showed similar sensitivity to IR as compared to normoxic HIF-1
knockdown U87 control cells, indicating that the increased radioresistance observed in
hypoxic U87 cells was mediated by HIF-1. The authors further confirmed these results
in vivo using a mouse xenograft model [89].
4.4. The Role of Glioma Stem Cells in Radioresistance
As early as 1997, Bonnet and Dick described for the first-time leukemic cells that
could transplant leukaemia
in vivo
into immunodeficient mice [
90
]. These cells were called
“cancer stem cells” (CSCs) and were later identified in different types of cancer including
Cancers 2021,13, 542 11 of 16
breast, colon, lung, as well as in CNS cancers such as GB [
91
]. CSCs are slow-dividing small
subpopulations of tumor cells that have the ability to undergo asymmetric cell division
for self-renewal and multilineage differentiation giving rise to more mature cancer cells
that constitute the bulk of the tumor [
92
,
93
]. The CSC hypothesis states that these cells
have the ability to generate the cellular heterogeneity which is commonly observed within
tumors. CSCs, called Glioma Stem Cells (GSCs) in GB, are suspected of being a main cause
of tumor recurrence after treatment.
GSCs have been shown to have similar properties to Neural Stem Cells (NSCs). In ad-
dition to self-renewal capacity and multilineage differentiation potential, GSCs also express
stemness markers involved in the regulation of specific signaling pathways, telomerase
activity, expression of ABC transporter proteins, migration, secretion of cytokines, growth
and pro-angiogenic factors [93].
It has been shown that hypoxia treatment of GB cells (e.g., SJ-1, U87) promoted
CD133 expression (marker for GSC) and increased OCT4 and SOX2 mRNA levels, while
promoting the loss of the glial differentiation marker, GFAP [
94
]. These data indicate
that hypoxia promotes a GSC phenotype in GB. Another report took a broader approach
by screening cell lines of different cancer types alongside human embryonic stem cells
for overlapping changes of common genes when grown under hypoxic conditions [
95
].
The authors showed that OCT4, NANOG, SOX2, KLF4, cMYC, and miRNA-302 were all
induced under hypoxic conditions in 11 different cancer cell lines from prostate, brain
(U251 GB cell line), kidney, cervix, lung, colon, liver and breast tumors [
95
]. This report
further supported a link between hypoxia and the stem cell phenotype by showing a
correlation between the expression of HIFs and OCT4 [95].
Whether HIF-1
α
, HIF-2
α
or both transcription factors play a main role in hypoxia
induced GSC phenotype is still a theme up for debate, with many studies showing differ-
ing results [29,9598].
It has been well established that CSCs are significantly more radioresistant as com-
pared to non-CSCs [
26
]. This is due to their enhanced DNA-repair capability, antioxidant
defenses (in particular ROS scavenging systems) and self-renewal potential [
99
]. Interest-
ingly, GSCs are found within a particular hypoxic environment, called a “niche”. This
hypoxic niche forces the GSCs to develop mechanisms of survival and resistance to this
harmful environment and also keeps the GSCs in a state of quiescence making them less
vulnerable to the effects of radiotherapy [
93
]. Upon radiotherapy, the GSC populations
containing advantageous genomic alterations that protect them against IR are selected and
continue to sustain the tumor leading to recurrence.
Hypoxia has been shown to promote the undifferentiated state of GSCs through the
activation of the Notch signaling pathway in a HIF dependent manner, contributing in
this manner to GB radioresistance [
100
]. In summary, hypoxia is able of inducing the
dedifferentiation of GB cells towards a GSCs phenotype, thus conferring aggressiveness
and increased resistance to radiotherapy.
Interestingly, Dahan et al. demonstrated that IR itself is capable of inducing ded-
ifferentiation of GB cells leading to overexpression of stem cell markers (SOX2, Nestin,
SHH, Nanog, EZH2, Olig2) and a decrease in glial and neuronal differentiation markers
(GFAP and OMgp, respectively) which resulted in increased tumorigenicity
in vivo
[
101
].
In addition, this team observed overexpression of survivin (a protein with anti-apoptotic
properties that is expressed during neurogenesis) after irradiation of GB cells. Inhibition of
this protein prevented the dedifferentiation of GB cells into GSCs [
101
]. This highlights a
potential signaling pathway involved in the dedifferentiation of GB cells and a prospective
therapeutic target which would make it possible to inhibit the acquisition of this stem-like
phenotype highly linked to GB recurrence.
5. Conclusions
Targeting hypoxia-mediated radioresistance is considered an attractive approach to
improve therapy outcome in GB. However, clinical trials evaluating the use of hypoxia-
Cancers 2021,13, 542 12 of 16
targeting agents have failed to reveal a benefit for GB patients, reviewed in [
102
]. This
emphasizes the need for more effective and mechanism-based therapies to overcome
hypoxia-induced radioresistance and the co-development of predictive biomarkers and
improved imaging of heterogeneous GB hypoxia to guide radiotherapy protocols. As
previously described, hypoxic areas of the tumor are more resistant to IR compared to
normoxic areas. For this reason, increased radiation doses or number of radiation cycles
within the GB hypoxic areas could be considered. Here we performed a review of the
current literature, regarding the molecular mechanisms which are activated by hypoxia
and that can potentially be targeted in the future to improve radiotherapy efficiency in GB
patients. We revealed that several signaling pathways involved in cell cycle regulation were
shown to provide radioresistance in hypoxic GB cells. These included the MEK/ERK/DNA-
PKc/HIF-1
α
; PI3K/AKT/mTORC1/4E-BP1; and inhibition of TCEAL7 transcription by
miR-301-a leading to activation of the Wnt/
β
-catenin signaling pathway. The glycolytic
metabolism which is triggered by hypoxia in GB cells was also shown to be involved in
radioresistance and its inhibition showed promising
in vivo
results using a mouse xenograft
model, providing radiosensitivity in the GB-bearing mice. These results are encouraging
regarding the development of glycolytic inhibiting therapy approaches in combination with
IR treatment. In addition, several ROS-dependent and Redox regulatory mechanisms were
shown to play a role in hypoxic GB radioresistance. These included enhanced expression
of GOT1 leading to increased levels of the antioxidant protein, GSH; up-regulation of
CIC which has been linked to mitochondrial membrane integrity and inhibition of ROS-
induced apoptosis; up-regulation of Nox4 and ROS-dependent up-regulation of HIF-1.
Taken together these data highlight that targeting key Redox systems in combination with
IR treatment might constitute a promising approach in GB therapy. Finally, the hypoxic
microenvironment has been shown to play a role in GSC maintenance and quiescence
which are associated with GB radioresistance. This occurs via inhibition of differentiation
markers (e.g., GFAP, OMgp) and induction of stemness markers (e.g., CD133, OCT4, SOX2,
NANOG, NESTIN, EZH2, SHH, KLF4, cMYC, Olig2).
In conclusion, targeting key cellular proteins/mechanisms involved in cell cycle regu-
lation, glycolytic metabolism, Redox regulation and/or GSC maintenance in combination
with IR treatment may potentially lead to the development of novel and effective therapies
for GB patients.
Funding:
P.A.M. was funded by an FCT Investigator contract from the Foundation for Science and
Technology (Fundação para a Ciência e a Tecnologia, FCT), Portugal (ref: IF/00614/2014) and FCT
exploratory grant, ref:IF/00614/2014/CP12340006. CBMR was financed by an FCT Research Center
Grant ref: UID/BIM/04773/2013CBMR1334. A.L.C. is supported by a PhD fellowship from the
University Paris Diderot.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Gilbert, M.R.; Wang, M.; Aldape, K.D.; Stupp, R.; Hegi, M.E.; Jaeckle, K.A.; Armstrong, T.S.; Wefel, J.S.; Won, M.; Blumenthal,
D.T.; et al. Dose-dense temozolomide for newly diagnosed glioblastoma: A randomized phase III clinical trial. J. Clin. Oncol.
2013,31, 4085–4091. [CrossRef] [PubMed]
2.
Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger,
K.; et al. 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. 2009,10, 459–466. [CrossRef] [PubMed]
3.
Wang, M.; Dignam, J.J.; Won, M.; Curran, W.; Mehta, M.; Gilbert, M.R. Variation over time and interdependence between disease
progression and death among patients with glioblastoma on RTOG 0525. Neuro-Oncology
2015
,17, 999–1006. [CrossRef] [PubMed]
4.
Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K.; Ellison, D.W. (Eds.) WHO Classification of Tumours of the Central Nervous
System WHO Classification of Tumours, 4th ed.; IARC Publications: Lyon, France, 2016. Available online: https://www.iarc.
who.int/news-events/iarc-publications-who-classification-of-tumours-of-the-central-nervous-system/ (accessed on 21 January
2021).
Cancers 2021,13, 542 13 of 16
5.
Nørøxe, D.S.; Poulsen, H.S.; Lassen, U. Hallmarks of glioblastoma: A systematic review. ESMO Open
2016
,1, e000144. [CrossRef]
[PubMed]
6.
McLendon, R.; Friedman, A.; Bigner, D.; Van Meir, E.G.; Brat, D.J.; Mastrogianakis, G.M.; Olson, J.J.; Mikkelsen, T.; Lehman, N.;
Aldape, K.; et al. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008,
455, 1061–1068. [CrossRef]
7.
Verhaak, R.G.W.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.;
et al. Cancer Genome Atlas Research Network Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma
characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010,17, 98–110. [CrossRef]
8.
Phillips, H.S.; Kharbanda, S.; Chen, R.; Forrest, W.F.; Soriano, R.H.; Wu, T.D.; Misra, A.; Nigro, J.M.; Colman, H.; Soroceanu, L.;
et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble
stages in neurogenesis. Cancer Cell 2006,9, 157–173. [CrossRef]
9.
Monteiro, A.R.; Hill, R.; Pilkington, G.J.; Madureira, P.A. The Role of Hypoxia in Glioblastoma Invasion. Cells
2017
,6, 45.
[CrossRef]
10.
Brennan, C.W.; Verhaak, R.G.W.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn,
J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013,155, 462–477. [CrossRef]
11.
Noushmehr, H.; Weisenberger, D.J.; Diefes, K.; Phillips, H.S.; Pujara, K.; Berman, B.P.; Pan, F.; Pelloski, C.E.; Sulman, E.P.; Bhat,
K.P.; et al. Cancer Genome Atlas Research Network Identification of a CpG island methylator phenotype that defines a distinct
subgroup of glioma. Cancer Cell 2010,17, 510–522. [CrossRef]
12.
Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.;
Bogdahn, U.; et al. European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups;
National Cancer Institute of Canada Clinical Trials Group Radiotherapy plus concomitant and adjuvant temozolomide for
glioblastoma. N. Engl. J. Med. 2005,352, 987–996. [CrossRef] [PubMed]
13.
Birzu, C.; French, P.; Caccese, M.; Cerretti, G.; Idbaih, A.; Zagonel, V.; Lombardi, G. Recurrent Glioblastoma: From Molecular
Landscape to New Treatment Perspectives. Cancers 2020,13, 47. [CrossRef] [PubMed]
14.
Yu, W.; Zhang, L.; Wei, Q.; Shao, A. O6-Methylguanine-DNA Methyltransferase (MGMT): Challenges and New Opportunities in
Glioma Chemotherapy. Front. Oncol. 2020,9, 1547. [CrossRef] [PubMed]
15.
Rajaratnam, V.; Islam, M.M.; Yang, M.; Slaby, R.; Ramirez, H.M.; Mirza, S.P. Glioblastoma: Pathogenesis and current status of
chemotherapy and other novel treatments. Cancers 2020,12, 937. [CrossRef]
16.
Yang, L.; Lin, C.; Wang, L.; Guo, H.; Wang, X. Hypoxia and hypoxia-inducible factors in glioblastoma multiforme progression
and therapeutic implications. Exp. Cell Res. 2012,318, 2417–2426. [CrossRef]
17.
Kaur, B.; Khwaja, F.W.; Severson, E.A.; Matheny, S.L.; Brat, D.J.; Van Meir, E.G. Hypoxia and the hypoxia-inducible-factor pathway
in glioma growth and angiogenesis. Neuro-Oncology 2005,7, 134–153. [CrossRef]
18.
Clara, C.A.; Marie, S.K.N.; de Almeida, J.R.W.; Wakamatsu, A.; Oba-Shinjo, S.M.; Uno, M.; Neville, M.; Rosemberg, S. Angiogenesis
and expression of PDGF-C, VEGF, CD105 and HIF-1αin human glioblastoma. Neuropathology 2014,34, 343–352. [CrossRef]
19.
Jensen, R.L.; Mumert, M.L.; Gillespie, D.L.; Kinney, A.Y.; Schabel, M.C.; Salzman, K.L. Preoperative dynamic contrast-enhanced
MRI correlates with molecular markers of hypoxia and vascularity in specific areas of intratumoral microenvironment and is
predictive of patient outcome. Neuro-Oncology 2014,16, 280–291. [CrossRef]
20. Kaplan, Y.S. Basic principles in radiation oncology. Cancer 1977,39 (Suppl. 2), 689–693. [CrossRef]
21.
Santivasi, W.L.; Xia, F. Ionizing Radiation-Induced DNA Damage, Response, and Repair. Antioxid. Redox Signal.
2014
,21, 251–259.
[CrossRef]
22.
Nickoloff, J.A.; Sharma, N.; Taylor, L. Clustered DNA Double-Strand Breaks: Biological Effects and Relevance to Cancer
Radiotherapy. Genes 2020,15, 99. [CrossRef] [PubMed]
23.
Le Caë, S. Water Radiolysis: Influence of Oxide Surfaces on H 2 Production under Ionizing Radiation. Water
2011
,3, 235–253.
[CrossRef]
24.
Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair and mutagenesis. Environ. Mol. Mutagen.
2017
,58, 235–263.
[CrossRef] [PubMed]
25.
Hill, R.; Leidal, A.M.; Madureira, P.A.; Gillis, L.D.; Waisman, D.M.; Chiu, A.; Lee, P.W.K. Chromium-mediated apoptosis:
Involvement of DNA-dependent protein kinase (DNA-PK) and differential induction of p53 target genes. DNA Repair
2008
,7,
1484–1499. [CrossRef] [PubMed]
26. Rycaj, K.; Tang, D.G. Cancer stem cells and radioresistance. Int. J. Radiat. Biol. 2014,90, 615–621. [CrossRef] [PubMed]
27.
Jordan, B.F.; Sonveaux, P. Targeting tumor perfusion and oxygenation to improve the outcome of anticancer therapy. Front.
Pharmacol. 2012,3, 94. [CrossRef] [PubMed]
28.
Pawlik, T.M.; Keyomarsi, K. Role of cell cycle in mediating sensitivity to radiotherapy. Int. J. Radiat. Oncol. Biol. Phys.
2004
,59,
928–942. [CrossRef]
29.
Bar, E.E.; Lin, A.; Mahairaki, V.; Matsui, W.; Eberhart, C.G. Hypoxia increases the expression of stem-cell markers and promotes
clonogenicity in glioblastoma neurospheres. Am. J. Pathol. 2010,177, 1491–1502. [CrossRef]
30.
Winkler, F.; Kozin, S.V.; Tong, R.T.; Chae, S.-S.; Booth, M.F.; Garkavtsev, I.; Xu, L.; Hicklin, D.J.; Fukumura, D.; di Tomaso, E.;
et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation,
angiopoietin-1, and matrix metalloproteinases. Cancer Cell 2004,6, 553–563. [CrossRef]
Cancers 2021,13, 542 14 of 16
31.
Yuan, F.; Salehi, H.A.; Boucher, Y.; Vasthare, U.S.; Tuma, R.F.; Jain, R.K. Vascular permeability and microcirculation of gliomas
and mammary carcinomas transplanted in rat and mouse cranial windows. Cancer Res. 1994,54, 4564–4568.
32.
Plate, K.H.; Mennel, H.D. Vascular morphology and angiogenesis in glial tumors. Exp. Toxicol. Pathol.
1995
,47, 89–94. [CrossRef]
33.
Bullitt, E.; Zeng, D.; Gerig, G.; Aylward, S.; Joshi, S.; Smith, J.K.; Lin, W.; Ewend, M.G. Vessel tortuosity and brain tumor
malignancy: A blinded study. Acad. Radiol. 2005,12, 1232–1240. [CrossRef] [PubMed]
34.
Rong, Y.; Durden, D.L.; Van Meir, E.G.; Brat, D.J. “Pseudopalisading” necrosis in glioblastoma: A familiar morphologic feature
that links vascular pathology, hypoxia, and angiogenesis. J. Neuropathol. Exp. Neurol. 2006,65, 529–539. [CrossRef] [PubMed]
35.
Jain, R.K.; di Tomaso, E.; Duda, D.G.; Loeffler, J.S.; Sorensen, A.G.; Batchelor, T.T. Angiogenesis in brain tumours. Nat. Rev.
Neurosci. 2007,8, 610–622. [CrossRef] [PubMed]
36.
Saxena, K.; Jolly, M.K. Acute vs. Chronic vs. cyclic hypoxia: Their differential dynamics, molecular mechanisms, and effects on
tumor progression. Biomolecules 2019,9, 339. [CrossRef]
37.
Majmundar, A.J.; Wong, W.J.; Simon, M.C. Hypoxia-Inducible Factors and the Response to Hypoxic Stress. Mol. Cell
2010
,40,
294–309. [CrossRef]
38.
Keith, B.; Johnson, R.S.; Simon, M.C. HIF1
α
and HIF2
α
: Sibling rivalry in hypoxic tumour growth and progression. Nat. Rev.
Cancer 2011,12, 9–22. [CrossRef]
39.
Huang, L.E.; Bunn, H.F. Hypoxia-inducible factor and its biomedical relevance. J. Biol. Chem.
2003
,278, 19575–19578. [CrossRef]
40. Schofield, C.J.; Ratcliffe, P.J. Oxygen sensing by HIF hydroxylases. Nat. Rev. Mol. Cell Biol. 2004,5, 343–354. [CrossRef]
41.
Semenza, G.L. Regulation of mammalian O2 homeostasis by hypoxia-inducible factor 1. Annu. Rev. Cell Dev. Biol.
1999
,15,
551–578. [CrossRef]
42.
Duan, C. Hypoxia-inducible factor 3 biology: Complexities and emerging themes. Am. J. Physiol. Cell Physiol.
2016
,310,
C260–C269. [CrossRef] [PubMed]
43.
Cockman, M.E.; Masson, N.; Mole, D.R.; Jaakkola, P.; Chang, G.-W.; Clifford, S.C.; Maher, E.R.; Pugh, C.W.; Ratcliffe, P.J.; Maxwell,
P.H. Hypoxia Inducible Factor-
α
Binding and Ubiquitylation by the von Hippel-Lindau Tumor Suppressor Protein. J. Biol. Chem.
2000,275, 25733–25741. [CrossRef] [PubMed]
44.
Maxwell, P.H.; Wiesener, M.S.; Chang, G.W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.E.; Wykoff, C.C.; Pugh, C.W.; Maher, E.R.;
Ratcliffe, P.J. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature
1999,399, 271–275. [CrossRef] [PubMed]
45.
Srinivas, V.; Zhang, L.P.; Zhu, X.H.; Caro, J. Characterization of an oxygen/redox-dependent degradation domain of hypoxia-
inducible factor alpha (HIF-alpha) proteins. Biochem. Biophys. Res. Commun. 1999,260, 557–561. [CrossRef]
46.
Ebert, B.L.; Bunn, H.F. Regulation of transcription by hypoxia requires a multiprotein complex that includes hypoxia-inducible
factor 1, an adjacent transcription factor, and p300/CREB binding protein. Mol. Cell. Biol. 1998,18, 4089–4096. [CrossRef]
47.
Huang, L.E.; Arany, Z.; Livingston, D.M.; Bunn, H.F. Activation of hypoxia-inducible transcription factor depends primarily
upon redox-sensitive stabilization of its alpha subunit. J. Biol. Chem. 1996,271, 32253–32259. [CrossRef]
48.
Kallio, P.J.; Pongratz, I.; Gradin, K.; McGuire, J.; Poellinger, L. Activation of hypoxia-inducible factor 1alpha: Posttranscriptional
regulation and conformational change by recruitment of the Arnt transcription factor. Proc. Natl. Acad. Sci. USA
1997
,94,
5667–5672. [CrossRef]
49.
Mahon, P.C.; Hirota, K.; Semenza, G.L. FIH-1: A novel protein that interacts with HIF-1alpha and VHL to mediate repression of
HIF-1 transcriptional activity. Genes Dev. 2001,15, 2675–2686. [CrossRef]
50.
Lando, D.; Peet, D.J.; Gorman, J.J.; Whelan, D.A.; Whitelaw, M.L.; Bruick, R.K. FIH-1 is an asparaginyl hydroxylase enzyme that
regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev. 2002,16, 1466–1471. [CrossRef]
51.
Zhang, P.; Yao, Q.; Lu, L.; Li, Y.; Chen, P.-J.; Duan, C. Hypoxia-inducible factor 3 is an oxygen-dependent transcription activator
and regulates a distinct transcriptional response to hypoxia. Cell Rep. 2014,6, 1110–1121. [CrossRef]
52.
Frederick, L.; Wang, X.Y.; Eley, G.; James, C.D. Diversity and frequency of epidermal growth factor receptor mutations in human
glioblastomas. Cancer Res. 2000,60, 1383–1387. [PubMed]
53.
Sansal, I.; Sellers, W.R. The biology and clinical relevance of the PTEN tumor suppressor pathway. J. Clin. Oncol.
2004
,22,
2954–2963. [CrossRef] [PubMed]
54.
Ravi, R.; Mookerjee, B.; Bhujwalla, Z.M.; Sutter, C.H.; Artemov, D.; Zeng, Q.; Dillehay, L.E.; Madan, A.; Semenza, G.L.; Bedi, A.
Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev.
2000
,14, 34–44.
[PubMed]
55.
Holland, E.C.; Hively, W.P.; DePinho, R.A.; Varmus, H.E. A constitutively active epidermal growth factor receptor cooperates
with disruption of G1 cell-cycle arrest pathways to induce glioma-like lesions in mice. Genes Dev.
1998
,12, 3675–3685. [CrossRef]
56.
Clarke, K.; Smith, K.; Gullick, W.J.; Harris, A.L. Mutant epidermal growth factor receptor enhances induction of vascular
endothelial growth factor by hypoxia and insulin-like growth factor-1 via a PI3 kinase dependent pathway. Br. J. Cancer
2001
,84,
1322–1329. [CrossRef]
57.
Zhong, H.; Chiles, K.; Feldser, D.; Laughner, E.; Hanrahan, C.; Georgescu, M.M.; Simons, J.W.; Semenza, G.L. Modulation of
hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP
pathway in human prostate cancer cells: Implications for tumor angiogenesis and therapeutics. Cancer Res. 2000,60, 1541–1545.
58.
Castaldo, S.A.; Freitas, J.R.; Conchinha, N.V.; Madureira, P.A. The Tumorigenic Roles of the Cellular REDOX Regulatory Systems.
Oxid. Med. Cell. Longev. 2016,2016, 8413032. [CrossRef]
Cancers 2021,13, 542 15 of 16
59.
Chédeville, A.L.; Lourdusamy, A.; Monteiro, A.R.; Hill, R.; Madureira, P.A. Investigating glioblastoma response to hypoxia.
Biomedicines 2020,8, 310. [CrossRef]
60.
Sanzey, M.; Abdul Rahim, S.A.; Oudin, A.; Dirkse, A.; Kaoma, T.; Vallar, L.; Herold-Mende, C.; Bjerkvig, R.; Golebiewska, A.;
Niclou, S.P. Comprehensive analysis of glycolytic enzymes as therapeutic targets in the treatment of glioblastoma. PLoS ONE
2015,10, e0123544. [CrossRef]
61.
Plate, K.H.; Breier, G.; Weich, H.A.; Risau, W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in
human gliomas in vivo. Nature 1992,359, 845–848. [CrossRef]
62.
Shweiki, D.; Itin, A.; Soffer, D.; Keshet, E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated
angiogenesis. Nature 1992,359, 843–845. [CrossRef]
63.
Arai, Y.; Kubota, T.; Nakagawa, T.; Kabuto, M.; Sato, K.; Kobayashi, H. Production of urokinase-type plasminogen activator
(u-PA) and plasminogen activator inhibitor-1 (PAI-1) in human brain tumours. Acta Neurochir.
1998
,140, 377–386. [CrossRef]
[PubMed]
64.
Kubala, M.H.; DeClerck, Y.A. The plasminogen activator inhibitor-1 paradox in cancer: A mechanistic understanding. Cancer
Metastasis Rev. 2019,38, 483–492. [CrossRef] [PubMed]
65.
Madureira, P.A.; Surette, A.P.; Phipps, K.D.; Taboski, M.A.S.; Miller, V.A.; Waisman, D.M. The role of the annexin A2 heterotetramer
in vascular fibrinolysis. Blood 2011,118, 4789–4797. [CrossRef] [PubMed]
66.
Madureira, P.A.; O’Connell, P.A.; Surette, A.P.; Miller, V.A.; Waisman, D.M. The biochemistry and regulation of S100A10: A
multifunctional plasminogen receptor involved in oncogenesis. J. Biomed. Biotechnol. 2012,2012, 353687. [CrossRef] [PubMed]
67.
Mongiardi, M.P.; Savino, M.; Falchetti, M.L.; Illi, B.; Bozzo, F.; Valle, C.; Helmer-Citterich, M.; Ferrè, F.; Nasi, S.; Levi, A. c-MYC
inhibition impairs hypoxia response in glioblastoma multiforme. Oncotarget 2016,7, 33257–33271. [CrossRef]
68.
Hu, Y.L.; DeLay, M.; Jahangiri, A.; Molinaro, A.M.; Rose, S.D.; Carbonell, W.S.; Aghi, M.K. Hypoxia-induced autophagy promotes
tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res. 2012,72, 1773–1783. [CrossRef]
69.
Tsai, Y.-T.; Wu, A.-C.; Yang, W.-B.; Kao, T.-J.; Chuang, J.-Y.; Chang, W.-C.; Hsu, T.-I. ANGPTL4 Induces TMZ Resistance of
Glioblastoma by Promoting Cancer Stemness Enrichment via the EGFR/AKT/4E-BP1 Cascade. Int. J. Mol. Sci.
2019
,20, 5625.
[CrossRef]
70.
Foltyn, M.; Luger, A.-L.; Lorenz, N.I.; Sauer, B.; Mittelbronn, M.; Harter, P.N.; Steinbach, J.P.; Ronellenfitsch, M.W. The phys-
iological mTOR complex 1 inhibitor DDIT4 mediates therapy resistance in glioblastoma. Br. J. Cancer
2019
,120, 481–487.
[CrossRef]
71.
Said, H.M.; Stein, S.; Hagemann, C.; Polat, B.; Staab, A.; Anacker, J.; Schoemig, B.; Theobald, M.; Flentje, M.; Vordermark,
D. Oxygen-dependent regulation of NDRG1 in human glioblastoma cells
in vitro
and
in vivo
.Oncol. Rep.
2009
,21, 237–246.
[CrossRef]
72. Said, H.M.; Safari, R.; Al-Kafaji, G.; Ernestus, R.I.; Löhr, M.; Katzer, A.; Flentje, M.; Hagemann, C. Time- and oxygen-dependent
expression and regulation of NDRG1 in human brain cancer cells. Oncol. Rep. 2017,37, 3625–3634. [CrossRef] [PubMed]
73.
Weiler, M.; Blaes, J.; Pusch, S.; Sahm, F.; Czabanka, M.; Luger, S.; Bunse, L.; Solecki, G.; Eichwald, V.; Jugold, M.; et al. mTOR
target NDRG1 confers MGMT-dependent resistance to alkylating chemotherapy. Proc. Natl. Acad. Sci. USA
2014
,111, 409–414.
[CrossRef] [PubMed]
74.
Luo, W.; Wang, Y. Hypoxia mediates tumor malignancy and therapy resistance. In Advances in Experimental Medicine and Biology;
Springer: New York, NY, USA, 2019; Volume 1136, pp. 1–18.
75.
Marampon, F.; Gravina, G.L.; Zani, B.M.; Popov, V.M.; Fratticci, A.; Cerasani, M.; Di Genova, D.; Mancini, M.; Ciccarelli, C.;
Ficorella, C.; et al. Hypoxia sustains glioblastoma radioresistance through ERKs/DNA-PKcs/HIF- 1
α
functional interplay. Int. J.
Oncol. 2014,45, 2121–2131. [CrossRef] [PubMed]
76.
Gouazé-Andersson, V.; Delmas, C.; Taurand, M.; Martinez-Gala, J.; Ene Evrard, S.; Mazoyer, S.; Toulas, C.; Cohen-Jonathan-Moyal,
E. Therapeutics, Targets, and Chemical Biology FGFR1 Induces Glioblastoma Radioresistance through the PLCg/Hif1a Pathway.
Cancer Res. 2016,76, 3036–3044. [CrossRef]
77.
De Young, M.P.; Horak, P.; Sofer, A.; Sgroi, D.; Ellisen, L.W. Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression
through REDD1-mediated 14-3-3 shuttling. Genes Dev. 2008,22, 239–251. [CrossRef]
78.
Pinto, J.A.; Rolfo, C.; Raez, L.E.; Prado, A.; Araujo, J.M.; Bravo, L.; Fajardo, W.; Morante, Z.D.; Aguilar, A.; Neciosup, S.P.; et al. In
silico evaluation of DNA Damage Inducible Transcript 4 gene (DDIT4) as prognostic biomarker in several malignancies. Sci. Rep.
2017,7, 1526. [CrossRef]
79.
Richter, J.D.; Sonenberg, N. Regulation of cap-dependent translation by eIF4E inhibitory proteins. Nature
2005
,433, 477–480.
[CrossRef]
80.
Dubois, L.; Magagnin, M.G.; Cleven, A.H.G.; Weppler, S.A.; Grenacher, B.; Landuyt, W.; Lieuwes, N.; Lambin, P.; Gorr, T.A.;
Koritzinsky, M.; et al. Inhibition of 4E-BP1 Sensitizes U87 Glioblastoma Xenograft Tumors to Irradiation by Decreasing Hypoxia
Tolerance. Int. J. Radiat. Oncol. Biol. Phys. 2009,73, 1219–1227. [CrossRef]
81.
Yue, X.; Lan, F.; Xia, T. Hypoxic Glioma Cell-Secreted Exosomal miR-301a Activates Wnt/
β
-catenin Signaling and Promotes
Radiation Resistance by Targeting TCEAL7. Mol. Ther. 2019,27, 1939–1949. [CrossRef]
82.
Shen, H.; Hau, E.; Joshi, S.; Dilda, P.J.; Mcdonald, K.L. Sensitization of Glioblastoma Cells to Irradiation by Modulating the
Glucose Metabolism. Mol. Cancer Ther. 2015,14, 1794–1804. [CrossRef]
Cancers 2021,13, 542 16 of 16
83.
Castaldo, S.A.; Ajime, T.; Serrão, G.; Anastácio, F.; Rosa, J.T.; Giacomantonio, C.A.; Howarth, A.; Hill, R.; Madureira, P.A. Annexin
A2 regulates akt upon h2o2-dependent signaling activation in cancer cells. Cancers 2019,11, 492. [CrossRef] [PubMed]
84.
Matschke, J.; Riffkin, H.; Klein, D.; Handrick, R.; Lüdemann, L.; Metzen, E.; Shlomi, T.; Stuschke, M.; Jendrossek, V. Targeted
Inhibition of Glutamine-Dependent Glutathione Metabolism Overcomes Death Resistance Induced by Chronic Cycling Hypoxia.
Antioxid. Redox Signal. 2016,25, 89–107. [CrossRef] [PubMed]
85.
Hlouschek, J.; Hansel, C.; Jendrossek, V.; Matschke, J. The mitochondrial citrate carrier (SLC25A1) sustains redox homeostasis
and mitochondrial metabolism supporting radioresistance of cancer cells with tolerance to cycling severe hypoxia. Front. Oncol.
2018,8, 170. [CrossRef] [PubMed]
86.
Peng, R.; Zhang, M.; Wang, H.; Lin, J.; Wang, H.; Wang, F.; Liu, L.; Zhao, Q.; Liu, J. Advances into Understanding the Vital Role of
the Mitochondrial Citrate Carrier (CIC) in Metabolic Diseases. Pharmacol. Res. 2020,161, 105132. [CrossRef] [PubMed]
87.
Catalina-Rodriguez, O.; Kolukula, V.K.; Tomita, Y.; Preet, A.; Palmieri, F.; Wellstein, A.; Byers, S.; Giaccia, A.J.; Glasgow, E.;
Albanese, C.; et al. The mitochondrial citrate transporter, CIC, is essential for mitochondrial homeostasis. Oncotarget
2012
,3,
1220–1235. [CrossRef] [PubMed]
88.
Hsieh, C.H.; Wu, C.P.; Lee, H.T.; Liang, J.A.; Yu, C.Y.; Lin, Y.J. NADPH oxidase subunit 4 mediates cycling hypoxia-promoted
radiation resistance in glioblastoma multiforme. Free Radic. Biol. Med. 2012,53, 649–658. [CrossRef]
89.
Hsieh, C.H.; Lee, C.H.; Liang, J.A.; Yu, C.Y.; Shyu, W.C. Cycling hypoxia increases U87 glioma cell radioresistance via ROS
induced higher and long-term HIF-1 signal transduction activity. Oncol. Rep. 2010,24, 1629–1636. [CrossRef]
90.
Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic
cell. Nat. Med. 1997,3, 730–737. [CrossRef]
91.
Crowder, S.W.; Balikov, D.A.; Hwang, Y.-S.; Sung, H.-J. Cancer Stem Cells Under Hypoxia as a Chemoresistance Factor in the
Breast and Brain. Curr. Pathobiol. Rep. 2014,2, 33–40. [CrossRef]
92. Jordan, C.T.; Guzman, M.L.; Noble, M. Cancer Stem Cells. N. Engl. J. Med. 2006,355, 1253–1261. [CrossRef]
93.
Fidoamore, A.; Cristiano, L.; Antonosante, A.; D’Angelo, M.; Di Giacomo, E.; Astarita, C.; Giordano, A.; Ippoliti, R.; Benedetti, E.;
Cimini, A. Glioblastoma Stem Cells Microenvironment: The Paracrine Roles of the Niche in Drug and Radioresistance. Stem Cells
Int. 2016,2016, 1–17. [CrossRef] [PubMed]
94.
Kolenda, J.; Jensen, S.S.; Aaberg-Jessen, C.; Christensen, K.; Andersen, C.; Brünner, N.; Kristensen, B.W. Effects of hypoxia on
expression of a panel of stem cell and chemoresistance markers in glioblastoma-derived spheroids. J. Neurooncol.
2011
,103, 43–58.
[CrossRef] [PubMed]
95.
Mathieu, J.; Zhang, Z.; Zhou, W.; Wang, A.J.; Heddleston, J.M.; Pinna, C.M.A.; Hubaud, A.; Stadler, B.; Choi, M.; Bar, M.; et al.
HIF induces human embryonic stem cell markers in cancer cells. Cancer Res. 2011,71, 4640–4652. [CrossRef] [PubMed]
96.
Seidel, S.; Garvalov, B.K.; Wirta, V.; Von Stechow, L.; Schänzer, A.; Meletis, K.; Wolter, M.; Sommerlad, D.; Henze, A.T.; Nistér, M.;
et al. A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2
α
.Brain
2010
,133, 983–995. [CrossRef]
[PubMed]
97.
Méndez, O.; Zavadil, J.; Esencay, M.; Lukyanov, Y.; Santovasi, D.; Wang, S.-C.C.; Newcomb, E.W.; Zagzag, D. Knock down of
HIF-1
α
in glioma cells reduces migration
in vitro
and invasion
in vivo
and impairs their ability to form tumor spheres. Mol.
Cancer 2010,9, 133. [CrossRef]
98.
Heddleston, J.M.; Wu, Q.; Rivera, M.; Minhas, S.; Lathia, J.D.; Sloan, A.E.; Iliopoulos, O.; Hjelmeland, A.B.; Rich, J.N. Hypoxia-
induced mixed-lineage leukemia 1 regulates glioma stem cell tumorigenic potential. Cell Death Differ.
2012
,19, 428–439. [CrossRef]
99.
Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells
promote radioresistance by preferential activation of the DNA damage response. Nature 2006,444, 756–760. [CrossRef]
100.
Rankin, E.B.; Giaccia, A.J. The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ.
2008
,15, 678–685. [CrossRef]
101.
Dahan, P.; Martinez Gala, J.; Delmas, C.; Monferran, S.; Malric, L.; Zentkowski, D.; Lubrano, V.; Toulas, C.; Cohen-Jonathan
Moyal, E.; Lemarie, A. Ionizing radiations sustain glioblastoma cell dedifferentiation to a stem-like phenotype through survivin:
Possible involvement in radioresistance. Cell Death Dis. 2014,5, e1543. [CrossRef]
102.
Bindra, R.S.; Chalmers, A.J.; Evans, S.; Dewhirst, M. GBM radiosensitizers: Dead in the water
. . .
or just the beginning? J.
Neurooncol. 2017,134, 513–521. [CrossRef]
... Borneol, a terpene from traditional Chinese medicine, sensitizes cells to TMZ by promoting HIF1α degradation, as demonstrated by Lin et al. [88]. Previous studies have also shown that borneol enhances the efficacy of doxorubicin [134], curcumin [135], cisplatin [136], and radiotherapy [137]. Liu et al. [79] demonstrated in preclinical studies the usefulness of mannose as an adjunct to TMZ and to enhance radiotherapy, and achieved long-term survival in mice. ...
... Developing combination therapies or innovative treatment strategies may be necessary to address this issue. Despite encouraging preclinical results, limited clinical data exist on the efficacy of HIF-related therapies in patients with GBM, necessitating further extensive clinical trials for validation [137,146]. Safety concerns, including potential side effects and toxicity, especially when combined with other treatments, require thorough evaluation [147][148][149]. ...
Article
Full-text available
Background: The study aims to investigate the role of hypoxia-inducible factors (HIFs) in the development, progression, and therapeutic potential of glioblastomas. Methodology: The study, following PRISMA guidelines, systematically examined hypoxia and HIFs in glioblastoma using MEDLINE (PubMed), Web of Science, and Scopus. A total of 104 relevant studies underwent data extraction. Results: Among the 104 studies, global contributions were diverse, with China leading at 23.1%. The most productive year was 2019, accounting for 11.5%. Hypoxia-inducible factor 1 alpha (HIF1α) was frequently studied, followed by hypoxia-inducible factor 2 alpha (HIF2α), osteopontin, and cavolin-1. Commonly associated factors and pathways include glucose transporter 1 (GLUT1) and glucose transporter 3 (GLUT3) receptors, vascular endothelial growth factor (VEGF), phosphoinositide 3-kinase (PI3K)-Akt-mechanistic target of rapamycin (mTOR) pathway, and reactive oxygen species (ROS). HIF expression correlates with various glioblastoma hallmarks, including progression, survival, neovascularization, glucose metabolism, migration, and invasion. Conclusion: Overcoming challenges such as treatment resistance and the absence of biomarkers is critical for the effective integration of HIF-related therapies into the treatment of glioblastoma with the aim of optimizing patient outcomes.
... Hypoxia-inducible factor 1 α (HIF-1α) is a transcription factor that activates multiple glioma survival signaling pathways [47,48]. Its level is increased by an oxygen concentration of about 2.5% to 10%, found in most glioblastoma multiforme [49]. ...
Preprint
Full-text available
Cancers, a large and heterogeneous group of malignancies, are becoming an increasingly important cause of premature mortality worldwide. 2% of these are primary CNS tumors, of which 24% are gliomas. These tumors are diverse, but what they have in common is one of the most unfavorable prognoses among all cancers. A patient's future varies depending on the grade of the diagnosed glioma. To date, however, no standard treatment for grade IV has been established. All glioblastomas eventually undergo progression or recurrence. The current standard of treatment, which includes surgical intervention, radiation therapy and chemotherapy, is therefore far from sufficient. Work is constantly underway to discover a new, effective form of glioma therapy. Photodynamic therapy (PDT) may be one of them. It involves the local or systemic application of a photosensitive compound - a photosensitizer (PS), which accumulates in the affected tissues. Photosensitizer molecules absorb light of the appropriate wavelength, initiating activation processes leading to the formation of reactive oxygen species and selective destruction of inappropriate cells. Currently, this method has been approved for the treatment of several cancers. Research focusing on the effective use of PDT in glioma therapy is already underway, with promising results. In our work, we present molecular insights into PDT of glioma. Based on the available literature, we analyze and systematize the impact of various molecules, proteins, transporters and transmitters on the efficacy of PDT and the effect of PDT on their expression. In addition, we highlight gaps in current knowledge and point out directions for future research that may contribute to the efficacy of PDT glioma.
... Hypoxia, which is often present in subregions of GBM tumours, is the leading cause of resistance to radiotherapy, is predictive of the local sites of recurrence and is associated with poor clinical outcomes in GBM (5)(6)(7)(8)(9)(10)(11)(12). Current standard of care treatment does not account for tumour hypoxia. ...
Article
Full-text available
Background Glioblastoma (GBM) is the most aggressive type of brain cancer, with a 5-year survival rate of ~5% and most tumours recurring locally within months of first-line treatment. Hypoxia is associated with worse clinical outcomes in GBM, as it leads to localized resistance to radiotherapy and subsequent tumour recurrence. Current standard of care treatment does not account for tumour hypoxia, due to the challenges of mapping tumour hypoxia in routine clinical practice. In this clinical study, we aim to investigate the role of oxygen enhanced (OE) and blood-oxygen level dependent (BOLD) MRI as non-invasive imaging biomarkers of hypoxia in GBM, and to evaluate their potential role in dose-painting radiotherapy planning and treatment response assessment. Methods The primary endpoint is to evaluate the quantitative and spatial correlation between OE and BOLD MRI measurements and [¹⁸F]MISO values of uptake in the tumour. The secondary endpoints are to evaluate the repeatability of MRI biomarkers of hypoxia in a test-retest study, to estimate the potential clinical benefits of using MRI biomarkers of hypoxia to guide dose-painting radiotherapy, and to evaluate the ability of MRI biomarkers of hypoxia to assess treatment response. Twenty newly diagnosed GBM patients will be enrolled in this study. Patients will undergo standard of care treatment while receiving additional OE/BOLD MRI and [¹⁸F]MISO PET scans at several timepoints during treatment. The ability of OE/BOLD MRI to map hypoxic tumour regions will be evaluated by assessing spatial and quantitative correlations with areas of hypoxic tumour identified via [¹⁸F]MISO PET imaging. Discussion MANGO (Magnetic resonance imaging of hypoxia for radiation treatment guidance in glioblastoma multiforme) is a diagnostic/prognostic study investigating the role of imaging biomarkers of hypoxia in GBM management. The study will generate a large amount of longitudinal multimodal MRI and PET imaging data that could be used to unveil dynamic changes in tumour physiology that currently limit treatment efficacy, thereby providing a means to develop more effective and personalised treatments.
... However, the resistance of GBM to TMZ can be as high as 50%, and the response to radiotherapy is often less than expected. This resistance may be attributed to metabolic alterations in GBM cells, the hypoxic tumor microenvironment, glioma stem cells, and the influence of miRNAs [25]. Moreover, NUCB2 knockdown does not affect MGMT expression, which suggests that NUCB2 might not be directly involved in the regulation of MGMT in GBM cells. ...
Article
Full-text available
Glioblastoma (GBM) stands as the most prevalent primary malignant brain tumor, typically resulting in a median survival period of approximately thirteen to fifteen months after undergoing surgery, chemotherapy, and radiotherapy. Nucleobindin-2 (NUCB2) is a protein involved in appetite regulation and energy homeostasis. In this study, we assessed the impact of NUCB2 expression on tumor progression and prognosis of GBM. We further evaluated the relationship between NUCB2 expression and the sensitivity to chemotherapy and radiotherapy in GBM cells. Additionally, we compared the survival of mice intracranially implanted with GBM cells. High NUCB2 expression was associated with poor prognosis in patients with GBM. Knockdown of NUCB2 reduced cell viability, migration ability, and invasion ability of GBM cells. Overexpression of NUCB2 resulted in reduced apoptosis following temozolomide treatment and increased levels of DNA damage repair proteins after radiotherapy. Furthermore, mice intracranially implanted with NUCB2 knockdown GBM cells exhibited longer survival compared to the control group. NUCB2 may serve as a prognostic biomarker for poor outcomes in patients with GBM. Additionally, NUCB2 not only contributes to tumor progression but also influences the sensitivity of GBM cells to chemotherapy and radiotherapy. Therefore, targeting NUCB2 protein expression may represent a novel therapeutic approach for the treatment of GBM.
Article
Background Hypoxia is associated with poor prognosis in many cancers including glioblastoma (GBM). Glioma stem-like cells (GSCs) often reside in hypoxic regions and serve as reservoirs for disease progression. Long non-coding RNAs (lncRNAs) have been implicated in GBM. However, the lncRNAs that modulate GSC adaptations to hypoxia are poorly understood. Identification of these lncRNAs may provide new therapeutic strategies to target GSCs under hypoxia. Methods lncRNAs induced by hypoxia in GSCs were identified by RNAseq. LUCAT1 expression was assessed by qPCR, RNAseq, Northern blot, single molecule FISH in GSCs, and interrogated in IvyGAP, TCGA, and CGGA databases. LUCAT1 was depleted by shRNA, CRISPR/Cas9, and CRISPR/Cas13d. RNAseq, Western blot, immunohistochemistry, co-IP, ChIP, ChIPseq, RNA immunoprecipitation, and proximity ligation assay were performed to investigate mechanisms of action of LUCAT1. GSC viability, limiting dilution assay, and tumorigenic potential in orthotopic GBM xenograft models were performed to assess the functional consequences of depleting LUCAT1. Results A new isoform of Lucat1 is induced by HIF1α and NRF2 in GSCs under hypoxia. LUCAT1 is highly expressed in hypoxic regions in GBM. Mechanistically, LUCAT1 formed a complex with HIF1α and its co-activator CBP to regulate HIF1α target gene expression and GSC adaptation to hypoxia. Depletion of LUCAT1 impaired GSC self-renewal. Silencing LUCAT1 decreased tumor growth and prolonged mouse survival in GBM xenograft models. Conclusions A HIF1α-LUCAT1 axis forms a positive feedback loop to amplify HIF1α signaling in GSCs under hypoxia. LUCAT1 promotes GSC self-renewal and GBM tumor growth. LUCAT1 is a potential therapeutic target in GBM.
Article
Full-text available
The tumor microenvironment (TME) is characterized by an acidic pH and low oxygen concentrations. Hypoxia induces neoplastic cell evasion of the immune surveillance, rapid DNA repair, metabolic reprogramming, and metastasis, mainly as a response to the hypoxic inducible factors (HIFs). Likewise, cancer cells increase matrix metalloproteinases’ (MMPs) expression in response to TME conditions, allowing them to migrate from the primary tumor to different tissues. Since HIFs and MMPs are augmented in the hypoxic TME, it is easy to consider that HIFs participate directly in their expression regulation. However, not all MMPs have a hypoxia response element (HRE)-HIF binding site. Moreover, different transcription factors and signaling pathways activated in hypoxia conditions through HIFs or in a HIF-independent manner participate in MMPs’ transcription. The present review focuses on MMPs’ expression in normal and hypoxic conditions, considering HIFs and a HIF-independent transcription control. In addition, since the hypoxic TME causes resistance to anticancer conventional therapy, treatment approaches using MMPs as a target alone, or in combination with other therapies, are also discussed.
Article
Full-text available
Simple Summary In this work, we focused on the synergistic antitumor effects of photodynamic therapy and ferroptosis. First, we briefly introduced the basic theory of ferroptosis and photodynamic therapy. We explored the synergistic anti-tumor effect of photodynamic therapy combined with ferroptosis from a mechanism perspective. Secondly, we introduced the application of photodynamic therapy combined with ferroptosis, which mainly includes the construction of nanomaterials and drug combination. Nanomaterials can exert synergistic effects by activating anti-tumor immunity, improving the hypoxic microenvironment, and inhibiting tumor angiogenesis. The drug combination strategy has good application prospects and clinical significance.We also discussed the shortcomings of existing combination treatment strategies and potential solutions. In conclusion, photodynamic therapy combined with ferroptosis is a promising combination anticancer strategy. Abstract Ferroptosis is a programmed death mode that regulates redox homeostasis in cells, and recent studies suggest that it is a promising mode of tumor cell death. Ferroptosis is regulated by iron metabolism, lipid metabolism, and intracellular reducing substances, which is the mechanism basis of its combination with photodynamic therapy (PDT). PDT generates reactive oxygen species (ROS) and ¹O2 through type I and type II photochemical reactions, and subsequently induces ferroptosis through the Fenton reaction and the peroxidation of cell membrane lipids. PDT kills tumor cells by generating excessive cytotoxic ROS. Due to the limited laser depth and photosensitizer enrichment, the systemic treatment effect of PDT is not good. Combining PDT with ferroptosis can compensate for these shortcomings. Nanoparticles constructed by photosensitizers and ferroptosis agonists are widely used in the field of combination therapy, and their targeting and biological safety can be improved through modification. These nanoparticles not only directly kill tumor cells but also further exert the synergistic effect of PDT and ferroptosis by activating antitumor immunity, improving the hypoxia microenvironment, and inhibiting the tumor angiogenesis. Ferroptosis-agonist-induced chemotherapy and PDT-induced ablation also have good clinical application prospects. In this review, we summarize the current research progress on PDT and ferroptosis and how PDT and ferroptosis promote each other.
Article
Glioblastoma is a tumor characterized by pronounced hypoxia. Hypoxia produces diverse effects on tumor cells, and the results of experimental studies available so far are contradictory. In vitro hypoxia can be modeled in two ways: by reducing the level of atmospheric oxygen (physically induced hypoxia) or by using hypoxia-inducing chemicals such as cobalt chloride (II) (CoCl2) (chemically induced hypoxia). In the present work, we analyzed the effect of CoCl2 on the viability, proliferation, and apoptosis of cells of three glioblastoma cell lines: 1321N1, T98g, and U373 MG. It was shown that CoCl2 induced a dose-dependent decrease in cell viability and proliferation, and at high concentrations (200 and 400 μM) stimulated cell death. CoCl2 had no effect on the cytotoxic activity of doxorubicin in two cell lines T98g and U373 MG, and enhanced the effect of the chemotherapeutic agent on the 1321N1 cell line, though no synergistic cytotoxic effect of the two agents was observed.
Article
Full-text available
Simple Summary Glioblastoma is the most common and aggressive malignant primary brain cancer in adults. The prognosis remains poor following standard-of-care treatment with surgery, radiotherapy and chemotherapy, with a median overall survival of about 15 months. Theoretically, all glioblastoma patients relapse. Once tumors progress after first-line therapy, treatment options are limited and management of recurrent glioblastoma remains challenging. In recent years, new treatments have been tested on recurrent glioblastoma patients. These include immunotherapy, antiangiogenic treatment, targeted therapy and combination regimens. Here, we review these treatment approaches and provide an overview on the molecular characteristics of recurrent glioblastoma. Abstract Glioblastoma is the most frequent and aggressive form among malignant central nervous system primary tumors in adults. Standard treatment for newly diagnosed glioblastoma consists in maximal safe resection, if feasible, followed by radiochemotherapy and adjuvant chemotherapy with temozolomide; despite this multimodal treatment, virtually all glioblastomas relapse. Once tumors progress after first-line therapy, treatment options are limited and management of recurrent glioblastoma remains challenging. Loco-regional therapy with re-surgery or re-irradiation may be evaluated in selected cases, while traditional systemic therapy with nitrosoureas and temozolomide rechallenge showed limited efficacy. In recent years, new clinical trials using, for example, regorafenib or a combination of tyrosine kinase inhibitors and immunotherapy were performed with promising results. In particular, molecular targeted therapy could show efficacy in selected patients with specific gene mutations. Nonetheless, some molecular characteristics and genetic alterations could change during tumor progression, thus affecting the efficacy of precision medicine. We therefore reviewed the molecular and genomic landscape of recurrent glioblastoma, the strategy for clinical management and the major phase I-III clinical trials analyzing recent drugs and combination regimens in these patients.
Article
Full-text available
Glioblastoma (GB) is the most common and deadly type of primary malignant brain tumor with an average patient survival of only 15–17 months. GBs typically have hypoxic regions associated with aggressiveness and chemoresistance. Using patient derived GB cells, we characterized how GB responds to hypoxia. We noted a hypoxia-dependent glycolytic switch characterized by the up-regulation of HK2, PFKFB3, PFKFB4, LDHA, PDK1, SLC2A1/GLUT-1, CA9/CAIX, and SLC16A3/MCT-4. Moreover, many proangiogenic genes and proteins, including VEGFA, VEGFC, VEGFD, PGF/PlGF, ADM, ANGPTL4, and SERPINE1/PAI-1 were up-regulated during hypoxia. We detected the hypoxic induction of invasion proteins, including the plasminogen receptor, S100A10, and the urokinase plasminogen activator receptor, uPAR. Furthermore, we observed a hypoxia-dependent up-regulation of the autophagy genes, BNIP-3 and DDIT4 and of the multi-functional protein, NDRG1 associated with GB chemoresistance; and down-regulation of EGR1 and TFRC (Graphical abstract). Analysis of GB patient cohorts’ revealed differential expression of these genes in patient samples (except SLC16A3) compared to non-neoplastic brain tissue. High expression of SLC2A1, LDHA, PDK1, PFKFB4, HK2, VEGFA, SERPINE1, TFRC, and ADM was associated with significantly lower overall survival. Together these data provide important information regarding GB response to hypoxia which could support the development of more effective treatments for GB patients.
Article
Full-text available
Glioblastoma is one of the most common and detrimental forms of solid brain tumor, with over 10,000 new cases reported every year in the United States. Despite aggressive multimodal treatment approaches, the overall survival period is reported to be less than 15 months after diagnosis. A widely used approach for the treatment of glioblastoma is surgical removal of the tumor, followed by radiotherapy and chemotherapy. While there are several drugs available that are approved by the Food and Drug Administration (FDA), significant efforts have been made in recent years to develop new chemotherapeutic agents for the treatment of glioblastoma. This review describes the molecular targets and pathogenesis as well as the current progress in chemotherapeutic development and other novel therapies in the clinical setting for the treatment of glioblastoma.
Article
Full-text available
Chemoresistance has been a significant problem affecting the efficacy of drugs targeting tumors for decades. MGMT, known as O6-methylguanine-DNA methyltransferase, is a DNA repair enzyme that plays an important role in chemoresistance to alkylating agents. Hence, MGMT is considered a promising target for tumor treatment. Several methods are employed to detect MGMT, each with its own advantages and disadvantages. Some of the detection methods are; immunohistochemistry, methylation-specific PCR (MSP), pyrophosphate sequencing, MGMT activity test, and real-time quantitative PCR. Methylation of MGMT promoter is a key predictor of whether alkylating agents can effectively control glioma cells. The prognostic value of MGMT in glioma is currently being explored. The expression of MGMT gene mainly depends on epigenetic modification–methylation of CpG island of MGMT promoter. CpG island covers a length of 762 bp, with 98 CpG sites located at the 5' end of the gene, ranging from 480 to 1,480 nucleotides. The methylation sites and frequencies of CpG islands vary in MGMT-deficient tumor cell lines, xenografts of glioblastoma and in situ glioblastoma. Methylation in some regions of promoter CpG islands is particularly associated with gene expression. The change in the methylation status of the MGMT promoter after chemotherapy, radiotherapy or both is not completely understood, and results from previous studies have been controversial. Several studies have revealed that chemotherapy may enhance MGMT expression in gliomas. This could be through gene induction or selection of high MGMT-expressing cells during chemotherapy. Selective survival of glioma cells with high MGMT expression during alkylating agent therapy may change MGMT status in case of recurrence. Several strategies have been pursued to improve the anti-tumor effects of temozolomide. These include the synthesis of analogs of O6-meG such as O6-benzylguanine (O6-BG) and O6-(4-bromothenyl) guanine (O6-BTG), RNAi, and viral proteins. This review describes the regulation of MGMT expression and its role in chemotherapy, especially in glioma. Targeting MGMT seems to be a promising approach to overcome chemoresistance. Further studies exploring new agents targeting MGMT with better curative effect and less toxicity are advocated. We anticipate that these developments will improve the current poor prognosis of glioma patients.
Article
Full-text available
Cells manage to survive, thrive, and divide with high accuracy despite the constant threat of DNA damage. Cells have evolved with several systems that efficiently repair spontaneous, isolated DNA lesions with a high degree of accuracy. Ionizing radiation and a few radiomimetic chemicals can produce clustered DNA damage comprising complex arrangements of single-strand damage and DNA double-strand breaks (DSBs). There is substantial evidence that clustered DNA damage is more mutagenic and cytotoxic than isolated damage. Radiation-induced clustered DNA damage has proven difficult to study because the spectrum of induced lesions is very complex, and lesions are randomly distributed throughout the genome. Nonetheless, it is fairly well-established that radiation-induced clustered DNA damage, including non-DSB and DSB clustered lesions, are poorly repaired or fail to repair, accounting for the greater mutagenic and cytotoxic effects of clustered lesions compared to isolated lesions. High linear energy transfer (LET) charged particle radiation is more cytotoxic per unit dose than low LET radiation because high LET radiation produces more clustered DNA damage. Studies with I-SceI nuclease demonstrate that nuclease-induced DSB clusters are also cytotoxic, indicating that this cytotoxicity is independent of radiogenic lesions, including single-strand lesions and chemically “dirty” DSB ends. The poor repair of clustered DSBs at least in part reflects inhibition of canonical NHEJ by short DNA fragments. This shifts repair toward HR and perhaps alternative NHEJ, and can result in chromothripsis-mediated genome instability or cell death. These principals are important for cancer treatment by low and high LET radiation.
Article
Full-text available
The paradoxical pro-tumorigenic function of plasminogen activator inhibitor 1 (PAI-1, aka Serpin E1) in cancer progression and metastasis has been the subject of an abundant scientific literature that has pointed to a pro-angiogenic role, a growth and migration stimulatory function, and an anti-apoptotic activity, all directed toward promoting tumor growth, cancer cell survival, and metastasis. With uPA, PAI-1 is among the most reliable biomarkers and prognosticators in many cancer types. More recently, a novel pro-tumorigenic function of PAI-1 in cancer-related inflammation has been demonstrated. These multifaceted activities of PAI-1 in cancer progression are explained by the complex structure of PAI-1 and its multiple functions that go beyond its anti-fibrinolytic and anti-plasminogen activation activities. However, despite the multiple evidences supporting a pro-tumorigenic role of PAI-1 in cancer, and the development of several inhibitors, targeting PAI-1, has remained elusive. In this article, the various mechanisms responsible for the pro-tumorigenic functions of PAI-1 are reviewed with emphasis on its more recently described contribution to cancer inflammation. The challenges of targeting PAI-1 in cancer therapy are then discussed.
Article
Full-text available
Glioblastoma (GBM) is the most aggressive type of brain tumor, with strong invasiveness and a high tolerance to chemotherapy. Despite the current standard treatment combining temozolomide (TMZ) and radiotherapy, glioblastoma can be incurable due to drug resistance. The existence of glioma stem-like cells (GSCs) is considered the major reason for drug resistance. However, the mechanism of GSC enrichment remains unclear. Herein, we found that the expression and secretion of angiopoietin-like 4 protein (ANGPTL4) were clearly increased in GSCs. The overexpression of ANGPTL4 induced GSC enrichment that was characterized by polycomb complex protein BMI-1 and SRY (sex determining region Y)-box 2 (SOX2) expression, resulting in TMZ resistance in GBM. Furthermore, epidermal growth factor receptor (EGFR) phosphorylation induced 4E-BP1 phosphorylation that was required for ANGPTL4-induced GSC enrichment. In particular, ANGPTL4 induced 4E-BP1 phosphorylation by activating phosphoinositide 3-kinase (PI3K)/AKT and extracellular signal–regulated kinase (ERK) cascades for inducing stemness. To elucidate the mechanism contributing to ANGPTL4 upregulation in GSCs, chromatin immunoprecipitation coupled with sequencing (ChIP-Seq) revealed that specificity protein 4 (Sp4) was associated with the promoter region, −979 to −606, and the luciferase reporter assay revealed that Sp4 positively regulated activity of the ANGPTL4 promoter. Moreover, both ANGPTL4 and Sp4 were highly expressed in GBM and resulted in a poor prognosis. Taken together, Sp4-mediated ANGPTL4 upregulation induces GSC enrichment through the EGFR/AKT/4E-BP1 cascade.
Article
Full-text available
Hypoxia has been shown to increase the aggressiveness and severity of tumor progression. Along with chronic and acute hypoxic regions, solid tumors contain regions of cycling hypoxia (also called intermittent hypoxia or IH). Cyclic hypoxia is mimicked in vitro and in vivo by periodic exposure to cycles of hypoxia and reoxygenation (H–R cycles). Compared to chronic hypoxia, cyclic hypoxia has been shown to augment various hallmarks of cancer to a greater extent: angiogenesis, immune evasion, metastasis, survival etc. Cycling hypoxia has also been shown to be the major contributing factor in increasing the risk of cancer in obstructive sleep apnea (OSA) patients. Here, we first compare and contrast the effects of acute, chronic and intermittent hypoxia in terms of molecular pathways activated and the cellular processes affected. We highlight the underlying complexity of these differential effects and emphasize the need to investigate various combinations of factors impacting cellular adaptation to hypoxia: total duration of hypoxia, concentration of oxygen (O2), and the presence of and frequency of H–R cycles. Finally, we summarize the effects of cycling hypoxia on various hallmarks of cancer highlighting their dependence on the abovementioned factors. We conclude with a call for an integrative and rigorous analysis of the effects of varying extents and durations of hypoxia on cells, including tools such as mechanism-based mathematical modelling and microfluidic setups.
Article
Because radiotherapy significantly increases median survival in patients with glioblastoma, the modulation of radiation resistance is of significant interest. High glycolytic states of tumor cells are known to correlate strongly with radioresistance; thus, the concept of metabolic targeting needs to be investigated in combination with radiotherapy. Metabolically, the elevated glycolysis in glioblastoma cells was observed postradiotherapy together with upregulated hypoxia-inducible factor (HIF)-1a and its target pyruvate dehydrogenase kinase 1 (PDK1). Dichloroacetate, aPDK inhibitor currently being used to treat lactic acidosis, can modify tumor metabolism by activating mitochondrial activity to force glycolytic tumor cells into oxidative phosphorylation. Dichloroacetate alone demonstrated modest antitumor effects in both in vitro and in vivo models of glioblastoma and has the ability to reverse the radiotherapy-induced glycolytic shift when given in combination. In vitro, an enhanced inhibition of clonogenicity of a panel of glioblastoma cells was observed when dichloroacetate was combined with radiotherapy. Further mechanistic investigation revealed that dichloroacetate sensitized glioblastoma cells to radiotherapy by inducing the cell-cycle arrest at the G2–M phase, reducing mitochondrial reserve capacity, and increasing the oxidative stress as well asDNAdamage in glioblastoma cells together with radiotherapy. In vivo, the combinatorial treatment of dichloroacetate and radiotherapy improved the survival of orthotopic glioblastoma-bearing mice. In conclusion, this study provides the proof of concept that dichloroacetate can effectively sensitize glioblastoma cells to radiotherapy by modulating the metabolic state of tumor cells. These findings warrant further evaluation of the combination of dichloroacetate and radiotherapy in clinical trials.
Article
The mitochondrial citrate carrier (CIC) is a nuclear-encoded protein located in the inner mitochondrial membrane. By mediating efflux of citrate from the mitochondria to the cytosol, CIC links mitochondrial central carbon metabolism and cytosolic lipogenesis together. Abnormal activity or expression of CIC was found in cancers, developmental disorders and many other diseases. Recently, the specific inhibitors of CIC were proved to modify basic cellular metabolism, which in turn led to changes in disease course such as reverted steatohepatitis and cancer cell death. CIC is believed to be a key player and may serve as a novel therapeutic target in types of human metabolic diseases. Therefore, in this paper, we integrally described the structure and function of CIC. Then, we gave an overview of CIC related diseases including cancers, congenital diseases, pro-inflammatory effects and some other diseases. At the same time, the potential molecular mechanisms of CIC in the above diseases were illuminated. Finally, we illuminated some emerging areas for future investigation.