ArticlePDF AvailableLiterature Review

Renin angiotensin system and its role in biomarkers and treatment in gliomas

Authors:

Abstract and Figures

Gliomas are the most common primary intrinsic tumor in the brain and are classified as low- or high-grade according to the World Health Organization (WHO). Patients with high-grade gliomas (HGG) who undergo surgical resection with adjuvant therapy have a mean overall survival of 15 months and 100% recurrence. The renin-angiotensin system (RAS), the primary regulator of cardiovascular circulation, exhibits local action and works as a paracrine system. In the context of this local regulation, the expression of RAS peptides and receptors has been detected in different kinds of tumors, including gliomas. The dysregulation of RAS components plays a significant role in the proliferation, angiogenesis, and invasion of these tumors, and therefore in their outcomes. The study and potential application of RAS peptides and receptors as biomarkers in gliomas could bring advantages against the limitations of current tumoral markers and should be considered in the future. The targeting of RAS components by RAS blockers has shown potential of being protective against cancer and improving immunotherapy. In gliomas, RAS blockers have shown a broad spectrum for beneficial effects and are being considered for use in treatment protocols. This review aims to summarize the background behind how RAS plays a role in gliomagenesis and explore the evidence that could lead to their use as biomarkers and treatment adjuvants.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Journal of Neuro-Oncology
https://doi.org/10.1007/s11060-018-2789-5
TOPIC REVIEW
Renin angiotensin system andits role inbiomarkers andtreatment
ingliomas
AlexanderPerdomo‑Pantoja1,6· SoniaIlianaMejía‑Pérez1· LilianaGómez‑Flores‑Ramos2,4·
MontserratLara‑Velazquez3,7· CordeliaOrillac5· JuanLuisGómez‑Amador1· TaliaWegman‑Ostrosky2
Received: 15 June 2017 / Accepted: 1 February 2018
© This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply 2018
Abstract
Gliomas are the most common primary intrinsic tumor in the brain and are classified as low- or high-grade according to the
World Health Organization (WHO). Patients with high-grade gliomas (HGG) who undergo surgical resection with adjuvant
therapy have a mean overall survival of 15months and 100% recurrence. The renin-angiotensin system (RAS), the primary
regulator of cardiovascular circulation, exhibits local action and works as a paracrine system. In the context of this local
regulation, the expression of RAS peptides and receptors has been detected in different kinds of tumors, includinggliomas.
The dysregulation of RAS components plays a significant role in the proliferation, angiogenesis, and invasion of these
tumors, and therefore in their outcomes. The study and potential application of RAS peptides and receptors as biomarkers
in gliomas could bring advantages against the limitations of current tumoral markers and should be considered in the future.
The targeting of RAS components by RAS blockers has shown potential of being protective against cancer and improving
immunotherapy. In gliomas, RAS blockers have shown a broad spectrum for beneficial effectsand are being considered for
use in treatment protocols. This review aims to summarize the background behind how RAS plays a role in gliomagenesis
and explore the evidence that could lead to their use as biomarkers and treatment adjuvants.
Keywords Glioblastoma· Renin-angiotensin system· Brain tumor· Glioma· Angiotensinogen· Cancer· Gliomagenesis
Introduction
Gliomas are the most common intrinsic neoplasm of the
brain and are derived from glial cells. Gliomas are classified
into astrocytomas, oligodendrogliomas, and ependymomas
depending on the glial cell-type of origin [1]. Among these,
astrocytomas are the most prevalent [2].
Gliomas are graded according to the World Health Organ-
ization (WHO) classification mainly based on their neuro-
pathologic features, with presence of cytological atypia con-
ferring a grade II, anaplasia and mitosis activity conferring
a grade III, and microvascular proliferation and/or necrosis
seen in grade IV tumors [35]. Grade I is reserved to the
more circumscribed pilocytic astrocytomas [3], or to an
extremely rare diffuse astrocytoma without atypia [5]. Grade
I and II gliomas are consider low-grade and grade III and IV
high-grade as a reflection of their respective growth rate and
malignant potential. High-grade gliomas are more common
than low-grade gliomas and are considered deadly, with a
worse prognosis despite the standard of care, and significant
morbidity [6]. Gliomas can also be grouped, based on the
* Talia Wegman-Ostrosky
taliaw@gmail.com
1 Department ofNeurological Surgery, National Institute
ofNeurology andNeurosurgery “Manuel Velasco Suarez”
(INNN), 3877 Insurgentes Sur Av, La Fama, Tlalpan,
14269MexicoCity, Mexico
2 Division ofResearch, National Cancer Institute
(INCAN), 22 San Fernando Av, Sección XVI, Tlalpan.,
14080MexicoCity, Mexico
3 School ofMedicine, Plan ofCombined Studies inMedicine
(PECEM), National Autonomous University ofMexico
(UNAM), MexicoCity, Mexico
4 Institute ofBiomedical Research, National Autonomous
University ofMexico (UNAM), MexicoCity, Mexico
5 NYU School ofMedicine, NYU Langone Health, NewYork,
USA
6 Department ofNeurosurgery, Johns Hopkins University
School ofMedicine, Baltimore, USA
7 Department ofNeurosurgery, Mayo Clinic Campus Florida,
Jacksonville, USA
Journal of Neuro-Oncology
1 3
WHO classification and the growth pattern, behavior, and
recently the isocitrate dehydrogenase enzyme 1 and 2 (IDH
1/2) genetic status [4]. Circumscribed tumors that maintain
well-defined limits are grade I, while grade II–IV tumors are
diffuse with borders that are difficult to distinguish from the
surrounding nerve tissue [4, 7]. Diffuse gliomas are thought
to be more of a systemic illness of the brain than a focal one
due to their infiltrating nature. Gross total resection (GTR)
of diffuse gliomas does not prevent recurrence [8, 9].
Diffuse gliomas have a high recurrence rate and a ten-
dency to progress from low-grade (grade II) to high-grade
tumors, either anaplastic grade III or even secondary glio-
blastomas (GBM) grade IV. For these tumors, the most
important neurosurgical objective is to perform an extensive
cytoreduction and obtain tumor tissue for diagnosis [10].
Due to their aggressive nature, GBMs are considered incura-
ble and have an adverse prognosis with significant morbidity.
For patients treated with GTR, adjuvant radiation therapy,
and adjuvant chemotherapy with temozolomide (TMZ), the
mean survival is 14.6months [11]. Since our current treat-
ments have limited mortality benefit, it is necessary to inves-
tigate new ways to approach the diagnosis and treatment of
gliomas, including the incorporation of molecular targets
into the management of gliomas [10]. There exist several
genes that have been proposed as biomolecular markers of
aggressive illness that provide clues of the pathophysiol-
ogy of brain tumors and illuminate some potential targets
for treatment [12]. In GBM, the majority of these markers
are detected directly from tumor tissue. In comparison, cir-
culating proteins in the blood seen in other types of cancer
(such as prostate-specific antigen in prostate cancer) serve
as markers of disease giving valuable information about the
differential diagnosis, prognosis, and response to treatment
in a less invasive manner [13].
In the past few years, ten mechanisms known as the “hall-
marks” of cancer have been proposed to explain the path
taken by tumor cells to acquire features that give them the
capacity to become malignant [14]. These hallmarks are
connected to several biochemical pathways that have been
the focus of much research, including the renin-angiotensin
system (RAS) [15]. Classically, the RAS has been studied as
a fundamental systemic component of cardiovascular home-
ostasis. However, RAS also is expressed in several tissues
and organs (liver, kidneys, pancreas, reproductive organs,
and brain), where it has paracrine regulation. Interestingly,
some of these local effects are related to carcinogenesis
including gliomagenesis [1618].
Epidemiologic studies have debated concerning the use of
antihypertensive drugs as protective agents against cancer,
and their role as neoadjuvant chemotherapy [15]. One of the
first studies to investigate the protective effect of RAS block-
ers on cancer in a clinical setting was a retrospective cohort
study based on 5207 patients, which found that the incidence
of fatal cancers was reduced in patients with long-term use
ofangiotensin-2 converting enzyme inhibitors (ACEIs) [19].
Going further, other epidemiological studies have combined
the genotype with the RAS blockers therapy, such as the
Rotterdam Study, a prospective cohort study with 7983 par-
ticipants with 1 of 4 cancers (colorectal, lung, breast, or
prostate cancer). The results showed that RAS inhibitors
seemed to protect against cancer in patients harboring the
ACE DD genotype [20]. Recently, Sun etal. published a
meta-analysis including 55 studies to evaluate the associa-
tion between RAS blockers and recurrence, metastasis and
survival in cancer patients. They found that those who used
RAS blockers had a longer progression-free survival (PFS)
and disease-free survival. In addition, it appears that the
positive influence of the RAS blockers in overall survival
(OS) depends on the cancer type and type of RAS inhibitor
used [21].
The role of RAS in CNS tumors has aroused increasing
interest among researchers in the field of neuro-oncology
over the past 20years. The purpose of this review is to
highlight the current knowledge concerning the relation-
ship between RAS and gliomagenesis, bearing in mind the
potential application of the RAS components as biomarkers
or treatment targets in cancer of the CNS.
The circulating RAS andthelocal RAS
The RAS plays an important action regulating the systemic
circulation in the human body in response to low blood pres-
sure or decrease in serum sodium levels. A component of
this model is angiotensinogen (AGT), a protein synthesized
and secreted by the liver into the general circulation. This
protein is converted through another step into angiotensin I
(AngI) by the protein renin from the renal juxtaglomerular
apparatus. Subsequently, AngI is converted to angiotensin
II (AngII) by the pulmonary angiotensin-converting enzyme
(ACE). AngII is an active octapeptide that acts primarily on
AngII receptor type 1 (AT1R). This promotes cardiovascular
homeostasis by upregulating the serum levels of aldosterone,
constricting blood vessels, and increasing salt reabsorption
and water retention at the level of the kidneys.
The new understanding of RAS is far more complex
than its classic view, and two important concepts have
been added. First, the system is not only expressed at a
systemic level but also works locally in a paracrine func-
tion in the vasculature, kidney, heart, lungs, liver and
central nervous system (CNS). Second, other peptides,
enzymes, and receptors have been added to the previous
framework of the RAS network, and include other bioac-
tive peptides in addition to AngII. Novel bioactive RAS
peptides discovered are angiotensin III (AngIII), angio-
tensin IV (AngIV), angiotensin (3–7), and angiotensin
Journal of Neuro-Oncology
1 3
(1–7), derived to a greater or lesser degree from AngII
[15, 22, 23]. Moreover, when renin releases Ang1 from
AGT, an additional large protein (des(Ang I)AGT) is
formed. Both AGT and des(Ang I)AGT are non-inhibitory
serpins and can inhibit angiogenesis [24]. (Fig.1).
The effects of the bioactive angiotensin peptides are
expressed mainly through AT1R and AT2R, but also
through the Mas receptor, the AngII receptor type 4
(AT4R), the insulin-regulated aminopeptidase (IRAP),
and ACE2 [22, 23, 25] (Fig.2). Furthermore, Ang-(1–7)
functions principally through the G protein-coupled
receptor Mas [26]. Currently, it is considered that ACE2/
Ang-(1–7)/Mas axis and AngII/ATR2 are antagonists of
the ACE/Ang II/AT1R axis, particularly under pathologi-
cal conditions [26, 27].
RAS inthebrain
In the early 1960s, AngII was found to be a neuroactive
peptide based on injections into dog heads that had a
central hypertensive response [28]. In the 1980s, it was
demonstrated that AngII does not penetrate the blood–brain
barrier (BBB) and consequently inferred that the brain AngII
does not come from systemic RAS [29]. Later, it was shown
that all essential substrates and enzymes needed for the syn-
thesis, metabolism, and action of the bioactive angiotensin
peptides can be produced locally in the brain, apart from
the peripheral system [30]. Nevertheless, all RAS compo-
nents have not been found in a single brain cell [31]. Fur-
thermore, the different brain RAS elements are distributed
in a heterogeneous and mismatched pattern, with some of
the components with a broad distribution and others with
restrained locations [32, 33]. This fact implies that the brain
RAS requires a complex network of intercellular interactions
to produce its bioactive neuropeptides, with the possibility
that biochemical or enzymatic pathways being used [31].
Peripheral angiotensins are capable of interacting with the
brain RAS at the circumventricular organs (CVO), structures
in the brain that represent a connection between the CNS and
the peripheral blood flow and are characterized by their large
vasculature and lack of a BBB [34]. It is considered that the
peripherally produced angiotensins play a significant role in
the control of certain behavioral, endocrine, and autonomic
Fig. 1 Scheme from left to right of AGT gene, mRNA, and Protein.
AGT gene is located on the long arm of chromosome 1 (1q42.3), with
five exons and four introns and 13 kilobases total (kb). AGT is a pro-
tein that contains 452 amino acids, classified as a member of the Ser-
pins family. AGT Angiotensinogen
Journal of Neuro-Oncology
1 3
functions through interactions at the CVOs. On the other
hand, the brain angiotensins perform central actions that are
not directly related to the systemic ones [3436].
In the brain, this central system has been linked to neu-
roinflammation, sensory information, learning and mem-
ory, neurodegeneration, neuroprotection, emotional stress
responses, and gliomagenesis [22].
Neuroinflammation is mainly regulated by the effects
of AngII; the activation of AT1R leads to inflammation,
increased oxidative stress, disruption of the BBB and finally
neurotoxicity. In addition, the activation of AT2R induces
DNA repair pathways and cellular differentiation [37]. It is
important to mention that the components of RAS are not
expressed homogeneously in the CNS. The AT1R and AT2R
share a comparable pattern of distribution, but not equal,
which has been seen in humans and other mammalians [31,
38]. Opposite to AT1R that prevail in adult species, AT2R is
highly expressed in the developing brain [31, 39, 40]. AT1R
is localized in high densities in areas related to neuroendo-
crine functions and cardiovascular autonomic regulation and
the limbic system [31]. These areas include the anterior pitu-
itary, area postrema, subfornical organ, median eminence,
lateral geniculate body, the solitary nucleus, inferior olivary
nucleus, the anterior ventral third ventricle region, and para-
ventricular, preoptic and supraoptic nuclei of the hypothala-
mus [22, 33]. Moreover, AT2R is found with higher densities
in the amygdala, caudate-putamen, hippocampus, thalamus,
globus pallidus, medial geniculate body, among others [22,
33].
On the other hand, AT4R has been found in basal ganglia,
cerebellum, anterior pituitary, neocortex, lateral geniculate
body, ventral lateral thalamic nucleus, motor cortex, and in
motor neurons of the brain stem and ventral horn of the
spinal cord [22, 37]. This pattern of distribution of AT4R
coincides with recent findings that suggest an essential role
of AngIV in memory enhancing and cognitive facilitation,
Fig. 2 Renin-angiotensin system in the brain. Peptides and enzymes
that participate in the conversion of angiotensinogen to angiotensin
I, as well as active forms and receptors. ACE angiotensin-converting
enzyme, ACE 2 angiotensin-converting enzyme 2, Ang III angiotensin
3, AT1R angiotensin II receptor type 1, AT2R angiotensin II receptor
type 2, MASR maspin receptor
Journal of Neuro-Oncology
1 3
effects that have been seen in response to ARBs [41]. Just
as the AT4R, the Mas receptor which is found in hippocam-
pus and amygdala, has been related to memory and learning
[37, 42]. Furthermore, the ACE2/Ang-(1–7)/Mas axis could
have an active and promising neuroprotective effect facing
ischemic stroke, as Ang-(1–7) increases the production of
both neuronal nitric oxide synthase (NOS) and endothelial
NOS [37].
Role ofRAS incancer
Analyzing the RAS under the Hanahan and Weinberg para-
digm of the tumorigenesis hallmarks gives an understanding
of how this system provides cancer cells with functional
capabilities that allow them to survive, proliferate and dis-
seminate [14]. Multiple experiments have demonstrated the
effect of RAS on sustaining proliferative signaling, evading
growth suppressors, resisting apoptosis, inducing angiogene-
sis, deregulating cellular energetics, as well as playing a role
in inflammation, cellular migration, invasion and metastasis
[14, 15, 18, 4350].
Distinct profiles of the gene expression of components of
RAS have been described in several types of cancers. For
example, the AGT gene has been associated with gastric can-
cer and H. Pylori, breast cancer in the post-menopause stage,
colorectal cancer, or renal cell carcinoma [5154]. Pringle
etal. showed that the G allele of AGT rs699 (g.9543T > C;
c.803T > C), which is associated with a 10–20% increase in
plasma AGT levels, was decreased in prevalence in women
with endometrial cancer. They also found that the AT1R sin-
gle nucleotide polymorphism (SNP) rs5186 (g.49331A > C;
c.*86A > C), which is known to be associated with over-
expression of the AT1R, is more prevalent in women with
endometrial cancer [55]. This same AT1R SNP and the SNP
rs1492078 (g.4520T > C; c.-869T > C) were studied in a
prospective cohort study in Netherlands. Having the AT1R
rs1492078 (g.4520T > C; c.-869T > C) polymorphism cor-
related with a decreased risk of developing renal cancer,
whereas AT1R rs5186 (g.49331A > C; c.*86A > C) showed
an increased risk. These associations were more significant
in patients with a prior history of hypertension [56].
Accumulating evidence has suggested that AT1R is
expressed in various tumors and that its expression is sig-
nificantly associated with tumor growth. The expression of
the AT1R receptor has been correlated with more aggressive
disease [18, 5759]. Arrieta etal. analyzed 77 breast tumors
and found that the expression of the AT1R receptor was
associated with a higher mitotic index, cellular proliferation
and angiogenesis [60].
AT1R and AT2R are G protein couple receptors with plei-
otropic activities with antagonistic effects. When activated,
AT1R promotes cellular proliferation and angiogenesis,
whereas AT2R has antiproliferative properties [15]. AT1R
activates several intracellular signaling pathways, includ-
ing second messengers PLCβ and Rho GEFs, and inositol
triphosphate, diacylglycerol and reactive oxygen species,
epidermal growth factor receptor (EGFR), platelet-derived
growth factor receptor (PDGFR), insulin-like growth fac-
tor-1 receptor (IGF-1R), G proteins including Ras, Rho
and Rac, and receptor activator of nuclear factor-κB ligand
(RANKL) [61]. Further, AngII leads to the upregulation of
transforming growth factor beta (TGF-β) in neuroinflamma-
tory processes, through the activation of AT1R and throm-
bospondin-1 (TSP-1) [62]. Importantly, AT1R signaling
leads to potent induction of vascular endothelial growth
factor (VEGF) [61]; the knowledge of this mechanism led
to the development of the interesting work of Levin etal.
where they describe a better survival in glioma patients
using angiotensin system inhibitors that were receiving
bevacizumab (BVZ) with/without cytotoxic chemotherapy
compared to those who were not using them [63]. While the
role of AT2R in cancer is less understood, it had been pos-
sible to see its effects. For instance, in an AT2R knockout
mice model, it was demonstrated that inhibition of AT2R
delays tumor growth by impairing VEGF expression [64].
Recently, antihypertensive drugs have been studied as
potential adjuvants in the treatment of cancer [18]. For
instance, a retrospective analysis of 269 bladder cancer
patients treated with ACEIs and angiotensin II receptor
blockers (ARBs) resulted with better overall survival (OS)
and cancer-specific survival (CSS) [65]. The effect of better
OS using ARBs has also been seen in patients with meta-
static breast and colorectal cancer by enhancing the effects
of BVZ [66]. Furthermore, the use of Ang-(1–7) inhibitors
is currently being studied in-vivo and in-vitro preclinical
models. Ang-(1–7) inhibitors work by obstructing cancer
cell growth, tumorigenesis, metastasis, and proliferation,
decreasing the expression of oncogenes, protein kinases and
cell cycle regulators, diminishing the expression of apopto-
sis inhibitors and promoting apoptosis, and also inhibiting
blood vessel formation in tumor cell lines. This has led to the
study of the use of Ang-(1–7) inhibitors as potential adju-
vants in chemotherapy and chemoprevention agents [67].
RAS ingliomagenesis andpotential
biomarkers
Several known clinical criteria serve as good prognostic
markers for gliomas such as younger age, maximal safe
resection, better Karnofsky Performance Score (KPS) and
lower WHO grade [3]. In the case of GBM, some biomark-
ers have shown to offer prognostic value. Of these mark-
ers, the IDH 1/2 mutation, the methylation of the promoter
Journal of Neuro-Oncology
1 3
O6-methylguanine-DNA methyltransferase (MGMT), and
the loss of heterozygosity (LOH) of 1p19q are used rou-
tinely for diagnostic and prognostic purposes, and they are
all considered good prognostic factors [68, 69]. Just as these
biomarkers are associated with carcinogenesis, the expres-
sion and regulation of RAS genes play a significant role in
the development and behavior of tumors, including gliomas.
This has been demonstrated both in-vivo and in-vitro, and
as a result, it has been hypothesized that the detection of
particular RAS components can lead to their application as
molecular biomarkers and that RAS antagonists could be
protective against cancer [15, 43].
The discovery of the RAS peptides and receptors in
GBM has encouraged their study from a different perspec-
tive. Indeed, in 2004, Juillerat-Jeanneret etal. reported that
AGT, prorenin, ACE, AT1R, and AT2R are produced and
expressed by human GBM and GBM cell in cultures. Also,
the presence of AGT in the pseudocyst of GBM, and ACE
expression in tumor-related vasculature were seen [17]. In
another study, Bradshaw etal. demonstrated that the com-
ponents of the RAS are also expressed in cancer stem cells
of GBMs [70]. Some specific RAS components have been
investigated in relation with gliomagenesis and have given
signs about how they could be applied as biomarkers, such
as the following.
AGT
In a retrospective study, predictor factors of BVZ response
were analyzed in recurrent GBM patients. An association
between the low and high expression of AGT and HLA class
II gene (human leukocyte antigen complex class II DQ alpha
1, HLA-DQA1) respectively, and a prolonged PFS and OS
in recurrent GBM patients treated with BVZ [71]. Further,
preliminary data from a prospective analysis performed by
our research team revealed a relation of the rs5050 AGT
polymorphism with prognosis in astrocytoma patients. In 48
patients harboring primary astrocytoma, a significant corre-
lation was found between the blood-detected GG-genotype
of rs5050 AGT and a lower survival rate (2 months in the
recessive group vs. 11 months in other groups, p 0.018) [72].
An extensive paper of the relation of several variants of the
AGT and gliomas is in progress [73].
ACE gene
Several studies have demonstrated a correlation between the
ACE I/D polymorphism and certain types of cancer [7480].
Recently, a Chinese population study identified the ACE DD
genotype as a risk factor for glioma. The DD genotype is
correlated with a higher plasma ACE activity in compari-
son to the other genotypes [81, 82]. This case-controlled
trial compared the ACE I/D genotypes between 800 glioma
patients and 800 controls, and it showed a higher presence
of ACE DD genotype in the glioma cases [83].
AT1R andAT2R
AT1R exhibits a range of effects in angiogenesis, cellular
proliferation, inflammation and cellular apoptosis as a posi-
tive regulator. In contrast, AT2R downregulates angiogen-
esis and inflammation [18]. This variability in biological
responses highlights the complexity of the role of RAS in
angiogenesis and cellular proliferation, either in a direct or
indirect way. These biological effects could suggest a link
between AT1R and AT2R and cancer in tumoral prolifera-
tion and angiogenesis. For instance, according to a transcrip-
tional network analysis, the positive expression of AT1R
and AT2R correlates with the regulation of different sets
of hub genes related to glioma progression. This hub-based
study reported that AT1R and AT2R inhibitions downregu-
late genes involved in protumoral functions in cells from
the C6 glioma cell line, suggesting both Ang-II receptors as
potential therapeutic targets [84].
A particular aspect to consider is the reliability of the
anti-AT1R antibodies for Western blot analysis and immu-
nohistochemistry. It has been reported the difficulty of pro-
ducing highly specific antibodies for G protein-coupled
receptors (such as AT1R), and a study performed in a mice
model demonstrated the lack of specificity of a commercial
panel of anti-AT1R antibodies [85]. Therefore, even though
AT1R isoforms have not been described in humans, it has to
be considered other quantitative or qualitative methods for
its detection (for instance, Northern blot or RT-PCR), either
for research or clinical purposes.
Ang‑(1–7)/Mas receptor
It has been demonstrated that some RAS components are
part of a counter balance axis that opposes AngII. Among
these counteractive RAS components are Ang-(1–7) and
the MAS receptor. This Ang-(1–7)/Mas axis plays an anti-
tumorigenesis role, decreasing growth and invasiveness in
some kinds of cancer [15, 86]. In-vitro, Ang-(1–7)/Mas sign-
aling inhibition by podocalyxin increased cell invasion and
proliferation in GBMs [87].
RAS blockers ingliomas
Antihypertensive drugs, in particular those that target
RAS components, like ACE inhibitors (ACEI) and AT1R
antagonists, have been studied as an emerging therapy to
affect tumor progression. So far, there are four known clus-
ters of RAS blockers: direct renin inhibitors (DRI) (i.e.
aliskiren), ACEIs (i.e. captopril, enalapril), AT1R blockers
Journal of Neuro-Oncology
1 3
(i.e. losartan) and aldosterone antagonists (i.e. spironolac-
tone) [88]. The use of approved antihypertensive drug to
treat cancer could be advantageous due to their availabil-
ity, good side effect profile, and low cost since they do not
need to go through preclinical studies and some early clini-
cal trials. However, outcomes are difficult to predict due to
the complexity of the RAS role in carcinogenesis [15, 67].
According to retrospectives analyses, antihypertensive drug
therapy has prophylactic and protective potential in suscep-
tible individuals against several types of cancer, including
prostate and breast [19, 8991].
In the context of gliomas there has been some works
that use RAS blockers (Table1). Rivera etal. published
the first study that demonstrated the presence of AT1R
on glioma cells, as well as the impact of the blockage of
AT1R with a losartan (a selective AT1R antagonist which
is able to cross the BBB) on the tumor development. In this
study, the authors inoculated glioma cells subcutaneously
in rats, which posteriorly were treated with different losar-
tan doses for 30days. Interestingly, the results showed a
dose-dependent change in the behavior of subcutaneous C6
rat gliomas using losartan. The higher dosage of losartan,
the greater decrease in tumoral volume, mitotic index, cell
proliferation, and the number of capillary vessels [16]. The
presence of angiogenic cytokines related to tumor angio-
genesis (VEGF, PDGF, bFGF, EGF, TNFα, and TGF α and
β) is insignificant in the brain under normal conditions [92].
However, the stimulation of AT1R in glioma can lead to
the overexpression of some of these growth-related factors,
and therefore, promote cell proliferation and angiogenesis.
It has been observed during glial tumorigenesis that AT1R
can induce the expression of PDGF, which in turn causes
liberation of VEGF [93]. This anti-angiogenic mechanism,
along with the increase of bioavailability of AngII for AT2R,
Table 1 Summary of reports RAS blockers in gliomas
AT1R angiotensin II type-1 receptor, ACE aldosteron converting enzyme, OS overall survival, PFS progression-free survival, KPS Karnofsky
performance status, RT radiotherapy, TMZ temozolamide, FLAIR fluid attenuated inversion recovery, MRI magnetic resonance imaging, BVZ
Bevacizumab
Author & year Antihypertensive group Antihypertensive drug Effect or outcomes on
gliomas
Type of assay
Rivera etal. 2001 [16] AT1R antagonist Losartan Tumor volume
Mitotic index
Cell proliferation
Vascular density
In vivo; subcutaneously
inoculated C6 rat glioma
cells implantation
Jiullerat-Jeanneret etal.
2000 [94]
ACE inhibitor Lisinopril No significance difference
in glioma features
In vivo; stereotactic trans-
plantation of the rat glioma
G2 cells in syngeneic rat
brains
Jiullerat-Jeanneret 2004
[17]
Renin-selective inhibitor RO0663525 DNA synthesis
Apoptosis
In vitro; human glioblastoma
cells in culture
Arrieta etal. (2005) [97] AT1R antagonist Losartan In vivo:
Tumor volume
Apoptotic rate
Growth factors
In vitro:
Cell viability
Apoptosis rate
In vivo; subcutaneously
inoculated C6 rat glioma
cells implantation. Invitro;
C6 glioma cells cultures
Carpentier etal. (2012)
[101]
ACE inhibitor & AT1R
antagonist
Not specified Steroid dosage required Clinical retrospective study;
GBM patients treated with
standard of care
Carpentier etal. (2015)
[100]
ACE inhibitor & AT1R
antagonist
Not specified OS
PFS
KPS Post-RT
Steroid doses
Clinical retrospective study;
GBM patients treated with
RT and TMZ
Carpentier etal. (2016)
[102]
AT1R Antagonist Not specified Volume of peri-tumoral
hyper T2-FLAIR signal
(decreased edema)
Cross sectional study; GBM
patients treated with ARBs
for high blood pressure,
with pre-operative MRI
without steroids
Levin etal. (2017) [63] ACE inhibitor & AT1R
antagonist
Not specified OS Clinical retrospective study;
Grade II–IV gliomas
patients treated with
chemotherapy and/or BEV
Journal of Neuro-Oncology
1 3
could explain the response of the inoculated gliomas to the
AT1R antagonist seen in this experiment.
One year before, Jiullerat-Jeanneret etal. conducted an
invitro and invivo study, where they demonstrated renin
immunoreactivity in both human brain parenchyma and
GBM cells, and high expression of ACE in tumor vascu-
lature. Their most significant finding was the higher ami-
nopeptidase A (APA) activity in tumor vessels, which was
demonstrated not to be related to neovascularization. This
APA overexpression was downregulated by TGFβ, infer-
ring a role of this peptide in the APA activity regulation in
endothelial cells. In the animal model used for evaluating the
invivo expression of APA and the effects of the angiotensins
blockage, they found that chronic intake of lisinopril did
not affect the glioma growth or size in their rat model. The
difference between the treated and non-treated groups was
not significant for glioma features, including tumor size and
APA activity in tumor vessels [94]. This study was interest-
ing in an additional sense because it proposed the APA as
a potential marker of chronic dysfunction of the metabolic
BBB, including loss of TGFβ capacity. It infers the accumu-
lation of AngIII in the tumor surroundings, instead of AngII
or AngIV which both own a significant role in the preserva-
tion of the normal BBB [95]. The anti-edema effect of the
glucocorticoids in brain tumor coincides with this finding,
as the steroids promote the ACE activity and diminish APA
expression, and therefore increasing AngII [95].
A subsequent invitro study by Jiullerat-Jeanneret etal.
evaluated the expression and function of the RAS peptides
and enzymes in GBM cell cultures. They found that AGT,
prorenin, ACE, AT1R, and AT2R are formed, and hetero-
geneously expressed, in human GBM and GBM cells in
culture. Furthermore, they detected AGT in the fluid from
human GBM pseudocyst, as well as ACE in tumor vessels
(as it was showed in their previous studies [94]). This sup-
ported the statement that all RAS components are potentially
found, but non-homogeneously, in the tumor environment.
The cell proliferation and survival were not affected by add-
ing AGT, des(Ang I)AGT, tetradecapeptide renin substrate
(AGT1-14), AngI, AngII or AngIII, directly to the GBM
cell cultures. Then, they evaluated the inhibition of the renin
and ACE enzymes. Cell proliferation was not altered with
the ACEI administration (captopril -thiol ACEI-, or lisino-
pril -non-thiol ACEI-). On the other hand, one of the renin
inhibitors tested, the RO0663525 (a synthetic renin-selective
inhibitor) decreased the DNA synthesis, the number of via-
ble tumor cells, and induced apoptosis (in cells previously
deprived of fetal calf serum), suggesting its potential use
with other drugs to delay tumoral progression [17]. This
difference in the effect of the renin inhibitors can be due
to the lipophilicity and structure of the inhibitor and to an
intracellular function of renin. The blockage of this intracel-
lular action of the renin could prevent the activation of ERK
pathways, which are involved in tumor survival signaling
[96].
In 2005, Arrieta etal. assessed the impact of selective
AT1R blockage with losartan on the synthesis of neoangio-
genesis-related growth factors and the induction of apoptosis
in C6 glioma rat subcutaneous tumor models and cultured
C6 glioma cells [97]. In the animal model, rats were given
an injection of C6 glioma cells, and subsequently developed
tumor subcutaneously at the inoculation site. Next, losartan
(40- or 80mg/kg−1) was administered orally, once a day, per
30days. The tumoral volume had a dose-dependent reduc-
tion with the use of losartan, greater in 80mg/kg−1 losar-
tan group, without a natural decrease in the control group.
Apoptotic rate was measured in these tumors, and only the
80mg/kg−1 losartan dose had a significant effect. Platelet-
derived growth factor (PDGF), basic fibroblast growth factor
(bFGF) and VEGF concentrations were significantly dimin-
ished as a result of treatment with Losartan. No difference
was seen on hepatocyte growth factor (HGF) between groups
[97]. It is remarkable that the effect of the AT1R blockage
on the reduced tumor growth coincides with the diminish-
ing of these angiogenic growth factors, mainly with PDGF.
The reduction of VEGF and bFGF was not related to losar-
tan dosage as it was to the reduction of PDGF; hence we
can infer that their reduction was due to the blockage of the
PDGF stimuli, instead of AT1R. Additionally, within the
first hours of administration of concomitant losartan and
AngII, glioma cell cultures showed a decline in cell viabil-
ity and increase in the rate of apoptosis, while when AngII
was given alone, a rise in cell proliferation was observed
[97]. This phenomenon might be caused by the existence of
ATR1 and ATR2 in the glioma cells and their dual effects
[16, 18, 98, 99].
In 2015, Carpentier etal. reported the first retrospec-
tive analysis that evaluated the effect of AngII inhibitors
on the clinical outcome in newly diagnosed GBM patients
treated with RT and TMZ with/without AngII inhibitors.
Prior to the commencement of the study, 73% of the AngII
inhibitors-treatment group had already been taking ARBs
and 27% had already been using ACEIs. They found that
the cohort treated with AngII inhibitors (ARBs or ACEIs)
had better outcomes in comparison to the control group.
These results were reflected in a significantly higher perfor-
mance status (better KPS) at one and six months post-RT,
as well as longer PFS and OS [100]. An intriguing point of
this study might be the gathering of both AngII inhibitors
(ARBs and ACEIs) in only one group. On the one hand,
the improvement of the functional and survival outcomes
observed in these GBM patients warrants further investiga-
tion with prospective trials; but on the other hand, it remains
unclear if a difference exists between the ARBs and ACEIs
effects in a clinical setting. As the pre-clinical studies that
we commented earlier can hint it, ARBs and ACEIs have
Journal of Neuro-Oncology
1 3
a discrepancy between their effects in glioma. ARBs have
shown to reduce the tumor growth, and the neoangiogenesis-
related growth factors (PDGF, VEGF, bFGF) expressed in
C6 glioma rat models [16, 97]; whilst the ACEIs, except
for renin-selective RO0663525, did not show an impact on
GBM cell proliferation [17, 94].
Concerning the glucocorticoid use, the AngII inhibitors-
treated group required a lower steroid dosage, consistent
with two previous studies of the same group, which reported
that RAS blockade is related to vasogenic edema reduc-
tion and steroid-sparing effect in GBM [101, 102]. In the
prospective and retrospective studies by Carpentier etal.,
from 2012 to 2016 respectively, which were focused on the
anti-edema effect of the AngII inhibitors, the ARBs were
examined independently of the ACEIs. The results revealed
a lower steroid-dosage needed [101], and peri-tumoral
edema reduction on MRI (on the T2-Fluid Attenuated Inver-
sion Recovery (FLAIR) and Apparent Diffusion Coefficient
(ADC) sequences) on the part of ARBs [102].
Particularly remarkable is the fact that the potential
anti-edema effect has been recently proposed for a broad
spectrum of inflammatory brain disorders (such as neuro-
degenerative disorders, stroke, affective disorders, radia-
tion-induced damage, and traumatic brain injury), because
of the excessive brain AT1R activity found in early stages
for these disorders [103, 104]. Indeed, an invivo study by
Zhang etal. describes that the AngII induces neuroinflam-
mation and provoke BBB breakdown via oxidative stress
[105]. Additionally, the benefit of the ARBs on the modula-
tion of the BBB has already been reported. Fleegal-DeMotta
etal. studied invitro the effect of AngII and Telmisartan
(an AT1R inhibitor) in brain microvessel endothelial cells
(MECs). They found that AngII was related to an increase
in 125I-albumin permeability in MECs. This effect did not
change with the addition of PD123, 319 (an AT2R blocker),
but it was hindered by the use of telmisartan, which reduced
the transcytotic and paracellular BBB MEC permeability
[106]. Moreover, the ARB-induced vascular normalization
also may be related to the inhibitory action of the AT1R
blockage against the VEGF-induced vascular leakage, as it
was seen in an ischemic animal model [107].
Recently, Levin etal. published a relevant retrospective
clinical analysis, which evaluated the impact on survival of
the ARBs in GBM patients (newly diagnosed and recur-
rent GBM) treated with chemotherapy and/or BVZ. Also in
this study, the patients treated either with ACEIs or AngII
inhibitors, were analyzed as the same ARBs group. The
authors concluded that the use of ARBs offers a meaningful
OS advantage in glioma patients, particularly in recurrent
GBM patients treated with low-dose BVZ (7.5mg/kg every
3–4 weeks) with ARBs exposure [63]. Some aspects of this
work are worth mentioning. GBMs overexpress VEGF and
other neovascularization factors, and hence, anti-angiogenic
therapies have been proposed as part of the treatment for
these tumors. In regards to this, BVZ (a humanized mono-
clonal anti-VEGF antibody), is the most studied anti-VEGF
drug on GBM. However, the results of BVZ are far from
being entirely satisfactory. According to an extensive review
by Lu-Emerson etal. clinical evidence has shown that,
despite promising results in phase II trials for the use of this
anti-VEGF drug in GBM [108], the results of the addition
of BVZ to RT and TMZ-based chemotherapy in randomized
phase III trials exhibited an increase in PFS only but did not
show a significant gain in OS [108110]. This information
supports the relevance of the results found by Levin etal. as
their combined treatment of BVZ and ARBs prolonged OS
in their patients. They also reported a more significant ben-
efit in recurrent GBM when it was given low-dose BVZ with
ARBs [63], which coincides with their previous work that
mentions a better outcome using BVZ at half the standard
dose for progressive or recurrent GBM [111].
A 2013 study by Lombardi etal. proposed antiangiogenic-
induced hypertension as a clinical biomarker of prognosis
in patients with recurrent GBM treated with antiangiogenic
drugs, either BVZ or sorafenib (a small inhibitor of sev-
eral tyrosine protein kinases, such as VEGFR and PDGFR).
In this retrospective analysis, they found that patients who
developed antiangiogenic-induced hypertension had a bet-
ter disease control rate, 6month PFS, and longer overall
survival [112]. It is noteworthy that the authors did not men-
tion the characteristics of the antihypertensive treatment (for
instance, type of drug: ARB, ACEI, or other), even though it
can be assumed, according to the methodology and results,
that all patients who exhibited hypertension started pre-
scription. Based on the new-found evidence, it may raise
the question of whether the antiangiogenic-induced hyper-
tension was the positive prognosis factor, or was the use
of antihypertensive drugs. Although theories had been pro-
posed about the mechanism by which antiangiogenic therapy
induces hypertension, it remains not entirely clear [113].
Nevertheless, it has been a matter of debate the use of ACEIs
to treat this condition. The report of some cases supports
the notion that ACEIs play a role as a counterbalance of the
antiangiogenic effect of BVZ and lessen its clinical activity
in solid tumors [114]. The reason for this response is related
to changes in the tumor microenvironment given by ACEIs;
it has been demonstrated that the chronic ACEI treatment
leads to the accumulation of bradykinin, substance P, and
N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP), which
may undermine the profits of the antiangiogenic drugs [115,
116].
Low blood perfusion in tumors is another aspect to take
account when clinicians treat them through systemic drugs.
Tumor hypoperfusion is caused mostly by the increase in
vascular permeability and vascular compression. The reduc-
tion in blood perfusion leads to a drop of oxygen diffusion,
Journal of Neuro-Oncology
1 3
creating a hypoxic tumor microenvironment, and hinder
drugs penetration. Stylianopoulos and Jain proposed a math-
ematical model to predict the optimal therapeutic strategy to
reach the vascular normalization and relief of solid stress,
according to the tumor microenvironment features, in order
to improve the oxygen and drug delivery [117]. In accord-
ance with this work, Jain etal. performed a study by which
they demonstrated that the use of Losartan inactivates can-
cer-associated fibroblasts (CAFs) and diminishes the produc-
tion of matrix components (such as hyaluronan and collagen)
that are responsible for the solid stress and blood vessels
compression in collagen-rich tumors, and therefore amelio-
rating the vascular perfusion [118]. This identified effect of
ARBs increases drug and oxygen delivery into the tumor
environment, by reducing intratumor hydrostatic pressure
and vascular compression, improving tumor perfusion, and
therefore, enhancing chemo- and oxygen-dependent radio-
therapy [118120]. It is noteworthy that at the beginning of
the experiments, the authors tested the effect of lisinopril
(ACEI) versus losartan (selective AT1R inhibitor) on the
matrix, to determine the function of angiotensin receptor
signaling in tumor fibrosis. Interestingly they found a lower
impact on desmoplasia with lisinopril, suggesting an anti-
desmoplasia effect of AT2R (as ACEIs block both AT1 and
AT2 receptors) [118].
The ACE/Ang II/AT1R axis contributes to an immuno-
suppressive tumor milieu by promoting desmoplasia, stim-
ulating abnormal vasculature formation, and modulating
proinflammatory factors and immune cells [121]. Evidence
shows that this immunosuppressive milieu limits the effects
of novel immune checkpoint blockers. Therefore, it has been
suggested that ARBs have the potential to enhance immu-
notherapy of GBM and other tumors [63, 121123]. ARBs
have shown to reduce infiltration of immunosuppressive cell
types, and augment delivery of intratumoral effector T cells
and immunotherapeutic drugs. This ARBs effect can poten-
tially allow lower dosage of immunotherapy and, in turn,
reduce the undesired side effects [121].
Besides the potential effect of increasing the tumor oxy-
gen delivery and radiosensitivity [120], the ARBs have also
been suggested as therapy for preventing radiation-induced
brain damage [104]. This topic has attracted attention in
multiple settings, ranging from nuclear power plant acci-
dents to space missions [124126]. So far, the mechanism
of reduction of radiation injury is not completely under-
stood, but it might be related to the indirect inhibition of
TGF-β (contributor to radiation-induced fibrosis) via AngII
blockage [125]. Evidence suggests that the use of ARBs can
prevent or mitigate the irradiation-induced cognitive impair-
ment. In two studies, Robbins etal. demonstrated in an irra-
diation rat model that the L-158,809 (AT1R inhibitor) [127]
and ramipril (ACEI) [128] given before, during, and after
fractioned whole-brain irradiation prevent the perirhinal
cortex-dependent cognitive impairment after irradiation.
The effect of Ramipril on the irradiation-induced reduction
in neurogenesis remains unclear [128, 129].
Due to the evidence given by all pre-clinical and clinical
studies on the positive effects of AngII inhibitors in gliomas,
new clinical trials are running nowadays with the purpose of
testing the potential beneficial role of these drugs in GBM.
For instance, the Captopril was proposed in 2013 as part of
the treatment protocol named Coordinated Undermining of
Survival Paths, CUSP9* in recurrent GBM patients. This
protocol consists of nine drugs (including captopril) added
concomitant to low continuous doses of TMZ in relapsed
GBM, to block 17 different GBM growth-enhancing path-
ways. Clinical outcomes regarding this protocol have not
yet been reported [130, 131]. For the same year, the Angio-
tensin II Receptor Blockers, Steroids, and Radiotherapy in
Glioblastoma (ASTER) trial was registered in France, which
is a multicenter, double-blinded, and randomized study.
Carpentier etal. will assess the clinical outcomes in GBM
patients undergone to the standard management (RT with
TMZ-based chemotherapy), treated with losartan versus Pla-
cebo. In the 2018 update, the study is ongoing, whilst the
recruitment has already been completed [132].
Conclusion
Our understanding of the RAS, a model that was considered
canonical for a long time, has evolved enormously in recent
years. In the same way, our learning about the mechanisms
and biochemical pathways related to gliomas has advanced
at a fast pace reaching new limits. Despite these facts, the
part of this iceberg of knowledge that remains under the
surface is still immense. The pursuit of a better understand-
ing of pathways and their complex networks has led us to
the discovery of a connection between this new concept of
RAS and gliomagenesis. The detection of RAS peptides and
receptors in gliomas has attracted the interest particularly
of neuroncologists, neurologists, and neurosurgeons due to
their potential application as molecular biomarkers or targets
for treatment. Advantages are clear, for instance, as biomark-
ers, some of the RAS components can be detected by blood
sample, and as targets for treatments, ARBs are widely avail-
able for a lower cost in comparison to other anti-neoplastic
agents. So far, the evidence is scarce to determine the com-
prehensive role of the RAS in the glioma pathophysiology
and immunology, but it is enough to encourage the design
of randomized clinical trials (such as CUSP9* and ASTER)
aimed to elucidate the impact of specific RAS components
and ARBs therapy in glioma patients.
Acknowledgements Special thanks to Alejandro Lafuente for his
invaluable help with the figures of this review.
Journal of Neuro-Oncology
1 3
Funding This project was funded by CONACYT
(Salud-2013-01-202720).
Compliance with ethical standards
Conflict of interest The authors declare that they have no conflict of
interest.
Research involving human participants and/or animals This article
does not contain any studies with human participants or animals per-
formed by any of the authors
Informed consent This article does not require informed consent.
References
1. Xavier-Magalhaes A, Nandhabalan M, Jones C, Costa BM (2013)
Molecular prognostic factors in glioblastoma: state of the art
and future challenges. CNS Oncol 2(6):495–510. https ://doi.
org/10.2217/cns.13.48
2. Ostrom QT, Gittleman H, Fulop J, Liu M, Blanda R, Kromer C,
Wolinsky Y, Kruchko C, Barnholtz-Sloan JS (2015) CBTRUS
statistical report: primary brain and central nervous system
tumors diagnosed in the United States in 2008–2012. Neuro
Oncol 17(Suppl 4):iv1–iv62. https ://doi.org/10.1093/neuon c/
nov18 9
3. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC,
Jouvet A, Scheithauer BW, Kleihues P (2007) The 2007 WHO
classification of tumours of the central nervous system. Acta
Neuropathol 114(2):97–109. https ://doi.org/10.1007/s0040
1-007-0243-4
4. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-
Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P,
Ellison DW (2016) The 2016 World Health Organization clas-
sification of tumors of the central nervous system: a summary.
Acta Neuropathol 131(6):803–820. https ://doi.org/10.1007/s0040
1-016-1545-1
5. Daumas-Duport C, Scheithauer B, O’Fallon J, Kelly P (1988)
Grading of astrocytomas. A simple and reproducible method.
Cancer 62(10):2152–2165
6. de Groot JF (2015) High-grade gliomas. Continuum Neuro Oncol
21(2):332–344. https ://doi.org/10.1212/01.CON.00004 64173
.58262 .d9
7. Komori T (2015) Pathology and genetics of diffuse gliomas in
adults. Neurol Med Chirurg 55(1):28–37. https ://doi.org/10.2176/
nmc.ra.2014-0229
8. Sahm F, Capper D, Jeibmann A, Habel A, Paulus W, Troost D,
von Deimling A (2012) Addressing diffuse glioma as a systemic
brain disease with single-cell analysis. Arch Neurol 69(4):523–
526. https ://doi.org/10.1001/archn eurol .2011.2910
9. Ferris SP, Hofmann JW, Solomon DA, Perry A (2017) Charac-
terization of gliomas: from morphology to molecules. Virchows
Arch. https ://doi.org/10.1007/s0042 8-017-2181-4
10. Aldape K, Zadeh G, Mansouri S, Reifenberger G, von Deim-
ling A (2015) Glioblastoma: pathology, molecular mechanisms
and markers. Acta Neuropathol 129(6):829–848. https ://doi.
org/10.1007/s0040 1-015-1432-1
11. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ,
Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau
P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K,
Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, Lacombe
D, Cairncross JG, Mirimanoff RO, European Organisation for R,
Treatment of Cancer Brain. Radiation Oncology T G, National
Cancer Institute of Canada Clinical Trials G (2009) Effects of
radiotherapy with concomitant and adjuvant temozolomide
versus radiotherapy alone on survival in glioblastoma in a ran-
domised phase III study: 5-year analysis of the EORTC-NCIC
trial. Lancet Oncol 10(5):459–466. https ://doi.org/10.1016/
S1470 -2045(09)70025 -7
12. Karsy M, Neil JA, Guan J, Mahan MA, Colman H, Jensen RL
(2015) A practical review of prognostic correlations of molecular
biomarkers in glioblastoma. Neurosurg Focus 38(3):E4. https ://
doi.org/10.3171/2015.1.FOCUS 14755
13. Preusser M (2014) Neuro-oncology: a step towards clinical blood
biomarkers of glioblastoma. Nat Rev Neurol 10(12):681–682.
https ://doi.org/10.1038/nrneu rol.2014.208
14. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next
generation. Cell 144(5):646–674. https ://doi.org/10.1016/j.
cell.2011.02.013
15. Wegman-Ostrosky T, Soto-Reyes E, Vidal-Millan S, Sanchez-
Corona J (2015) The renin-angiotensin system meets the
hallmarks of cancer. J Renin-Angiotensin-Aldosterone Syst
16(2):227–233. https ://doi.org/10.1177/14703 20313 49685 8
16. Rivera E, Arrieta O, Guevara P, Duarte-Rojo A, Sotelo J (2001)
AT1 receptor is present in glioma cells; its blockage reduces the
growth of rat glioma. Br J Cancer 85(9):1396–1399. https ://doi.
org/10.1054/bjoc.2001.2102
17. Juillerat-Jeanneret L, Celerier J, Chapuis Bernasconi C, Nguyen
G, Wostl W, Maerki HP, Janzer RC, Corvol P, Gasc JM (2004)
Renin and angiotensinogen expression and functions in growth
and apoptosis of human glioblastoma. Br J Cancer 90(5):1059–
1068. https ://doi.org/10.1038/sj.bjc.66016 46
18. Ager EI, Neo J, Christophi C (2008) The renin-angiotensin sys-
tem and malignancy. Carcinogenesis 29(9):1675–1684. https ://
doi.org/10.1093/carci n/bgn17 1
19. Lever AF, Hole DJ, Gillis CR, McCallum IR, McInnes GT,
MacKinnon PL, Meredith PA, Murray LS, Reid JL, Robertson
JW (1998) Do inhibitors of angiotensin-I-converting enzyme
protect against risk of cancer? Lancet 352(9123):179–184. https
://doi.org/10.1016/S0140 -6736(98)03228 -0
20. van der Knaap R, Siemes C, Coebergh JW, van Duijn CM, Hof-
man A, Stricker BH (2008) Renin-angiotensin system inhibitors,
angiotensin I-converting enzyme gene insertion/deletion poly-
morphism, and cancer: the Rotterdam Study. Cancer 112(4):748–
757. https ://doi.org/10.1002/cncr.23215
21. Sun H, Li T, Zhuang R, Cai W, Zheng Y (2017) Do renin-angi-
otensin system inhibitors influence the recurrence, metastasis,
and survival in cancer patients?: evidence from a meta-analy-
sis including 55 studies. Medicine 96(13):e6394. https ://doi.
org/10.1097/MD.00000 00000 00639 4
22. Wright JW, Harding JW (2011) Brain renin-angiotensin–a new
look at an old system. Prog Neurobiol 95(1):49–67. https ://doi.
org/10.1016/j.pneur obio.2011.07.001
23. Crowley SD, Coffman TM (2012) Recent advances involving the
renin-angiotensin system. Exp Cell Res 318(9):1049–1056. https
://doi.org/10.1016/j.yexcr .2012.02.023
24. Celerier J, Cruz A, Lamande N, Gasc JM, Corvol P (2002)
Angiotensinogen and its cleaved derivatives inhibit angiogenesis.
Hypertension 39(2):224–228
25. Albiston AL, Fernando RN, Yeatman HR, Burns P, Ng L, Das-
wani D, Diwakarla S, Pham V, Chai SY (2010) Gene knockout
of insulin-regulated aminopeptidase: loss of the specific binding
site for angiotensin IV and age-related deficit in spatial memory.
Neurobiol Learn Mem 93(1):19–30. https ://doi.org/10.1016/j.
nlm.2009.07.011
26. Santos RA, Ferreira AJ, Verano-Braga T, Bader M (2013)
Angiotensin-converting enzyme 2, angiotensin-(1–7) and
Journal of Neuro-Oncology
1 3
Mas: new players of the renin-angiotensin system. J Endocrinol
216(2):R1–R17. https ://doi.org/10.1530/JOE-12-0341
27. Xu P, Sriramula S, Lazartigues E (2011) ACE2/ANG-(1–7)/
Mas pathway in the brain: the axis of good. Am J Physiol
Regul Integr Comp Physiol 300(4):R804-817. https ://doi.
org/10.1152/ajpre gu.00222 .2010
28. Bickerton RK, Buckley JP (1961) Evidence for a central
mechanism in angiotensin induced hypertension. Proc Soc
Exp Biol Med 106(4):834–836. https ://doi.org/10.3181/00379
727-106-26492
29. Harding JW, Sullivan MJ, Hanesworth JM, Cushing LL, Wright
JW (1988) Inability of [125I]Sar1, Ile8-angiotensin II to move
between the blood and cerebrospinal fluid compartments. J
Neurochem 50(2):554–557
30. Wright JW, Harding JW (1994) Brain angiotensin receptor sub-
types in the control of physiological and behavioral responses.
Neurosci Biobehav Rev 18(1):21–53
31. Saavedra JM (2005) Brain angiotensin II: new developments,
unanswered questions and therapeutic opportunities. Cell Mol
Neurobiol 25(3–4):485–512. https ://doi.org/10.1007/s1057
1-005-4011-5
32. Davisson RL (2003) Physiological genomic analysis of the
brain renin-angiotensin system. Am J Physiol Regul Integr
Comp Physiol 285(3):R498-511. https ://doi.org/10.1152/ajpre
gu.00190 .2003
33. und Halbach OV, Albrecht D (2006) The CNS renin-angio-
tensin system. Cell Tissue Res 326(2):599–616. https ://doi.
org/10.1007/s0044 1-006-0190-8
34. Fry M, Ferguson AV (2007) The sensory circumventricular
organs: brain targets for circulating signals controlling inges-
tive behavior. Physiol Behav 91(4):413–423. https ://doi.
org/10.1016/j.physb eh.2007.04.003
35. Ferguson AV, Washburn DLS, Bains JS (1999) Regulation of
autonomic pathways by angiotensin. Curr Opin Endocrinol
Diabet Obes 6(1):19
36. Ferguson AV, Washburn DL, Latchford KJ (2001) Hormonal
and neurotransmitter roles for angiotensin in the regulation of
central autonomic function. Exp Biol Med 226(2):85–96
37. Farag E, Sessler DI, Ebrahim Z, Kurz A, Morgan J, Ahuja S,
Maheshwari K, John Doyle D (2017) The renin angiotensin
system and the brain: New developments. J Clin Neurosci.
https ://doi.org/10.1016/j.jocn.2017.08.055
38. Barnes JM, Steward LJ, Barber PC, Barnes NM (1993) Identifi-
cation and characterisation of angiotensin II receptor subtypes
in human brain. Eur J Pharmacol 230(3):251–258
39. Tsutsumi K, Saavedra JM (1991) Characterization and develop-
ment of angiotensin II receptor subtypes (AT1 and AT2) in rat
brain. Am J Physiol 261(1 Pt 2):R209-216
40. Johren O, Saavedra JM (1996) Gene expression of angiotensin
II receptor subtypes in the cerebellar cortex of young rats.
Neuroreport 7(8):1349–1352
41. Wright JW, Harding JW (2013) The brain renin-angiotensin
system: a diversity of functions and implications for CNS dis-
eases. Pflugers Arch 465(1):133–151. https ://doi.org/10.1007/
s0042 4-012-1102-2
42. Lazaroni TL, Raslan AC, Fontes WR, de Oliveira ML, Bader
M, Alenina N, Moraes MF, Dos Santos RA, Pereira GS (2012)
Angiotensin-(1–7)/Mas axis integrity is required for the expres-
sion of object recognition memory. Neurobiol Learn Mem
97(1):113–123. https ://doi.org/10.1016/j.nlm.2011.10.003
43. George AJ, Thomas WG, Hannan RD (2010) The renin-angio-
tensin system and cancer: old dog, new tricks. Nat Rev Cancer
10(11):745–759. https ://doi.org/10.1038/nrc29 45
44. Vincent F, Bonnin P, Clemessy M, Contreres JO, Lamande N,
Gasc JM, Vilar J, Hainaud P, Tobelem G, Corvol P, Dupuy E
(2009) Angiotensinogen delays angiogenesis and tumor growth
of hepatocarcinoma in transgenic mice. Cancer Res 69(7):2853–
2860. https ://doi.org/10.1158/0008-5472.CAN-08-2484
45. Arrieta O, Pineda-Olvera B, Guevara-Salazar P, Hernandez-
Pedro N, Morales-Espinosa D, Ceron-Lizarraga TL, Gonzalez-
De la Rosa CH, Rembao D, Segura-Pacheco B, Sotelo J (2008)
Expression of AT1 and AT2 angiotensin receptors in astrocyto-
mas is associated with poor prognosis. Br J Cancer 99(1):160–
166. https ://doi.org/10.1038/sj.bjc.66044 31
46. Li JM, Mogi M, Tsukuda K, Tomochika H, Iwanami J, Min LJ,
Nahmias C, Iwai M, Horiuchi M (2007) Angiotensin II-induced
neural differentiation via angiotensin II type 2 (AT2) receptor-
MMS2 cascade involving interaction between AT2 receptor-
interacting protein and Src homology 2 domain-containing
protein-tyrosine phosphatase 1. Mol Endocrinol 21(2):499–511.
https ://doi.org/10.1210/me.2006-0005
47. Li H, Qi Y, Li C, Braseth LN, Gao Y, Shabashvili AE, Katovich
MJ, Sumners C (2009) Angiotensin type 2 receptor-medi-
ated apoptosis of human prostate cancer cells. Mol Cancer
Ther 8(12):3255–3265. https ://doi.org/10.1158/1535-7163.
MCT-09-0237
48. Bouquet C, Lamande N, Brand M, Gasc JM, Jullienne B, Faure
G, Griscelli F, Opolon P, Connault E, Perricaudet M, Corvol P
(2006) Suppression of angiogenesis, tumor growth, and metasta-
sis by adenovirus-mediated gene transfer of human angiotensino-
gen. Mol Ther 14(2):175–182. https ://doi.org/10.1016/j.ymthe
.2006.01.017
49. Smith GR, Missailidis S (2004) Cancer, inflammation and
the AT1 and AT2 receptors. J Inf lamm 1(1):3. https ://doi.
org/10.1186/1476-9255-1-3
50. Rodriguez A, Gomez-Ambrosi J, Catalan V, Fortuno A, Fruhbeck
G (2010) Leptin inhibits the proliferation of vascular smooth
muscle cells induced by angiotensin II through nitric oxide-
dependent mechanisms. Mediators Inflamm 2010:105489. https
://doi.org/10.1155/2010/10548 9
51. Gonzalez-Zuloeta Ladd AM, Arias Vasquez A, Siemes C,
Yazdanpanah M, Coebergh JW, Hofman A, Stricker BH, van
Duijn CM (2007) Differential roles of angiotensinogen and
angiotensin receptor type 1 polymorphisms in breast cancer
risk. Breast Cancer Res Treat 101(3):299–304. https ://doi.
org/10.1007/s1054 9-006-9290-0
52. Vasku A, Vokurka J, Bienertova-Vasku J (2009) Obesity-related
genes variability in Czech patients with sporadic colorectal can-
cer: preliminary results. Int J Colorectal Dis 24(3):289–294. https
://doi.org/10.1007/s0038 4-008-0553-6
53. Sugimoto M, Furuta T, Shirai N, Kodaira C, Nishino M, Ikuma
M, Sugimura H, Hishida A (2007) Role of angiotensinogen gene
polymorphism on Helicobacter pylori infection-related gastric
cancer risk in Japanese. Carcinogenesis 28(9):2036–2040. https
://doi.org/10.1093/carci n/bgm07 4
54. Andreotti G, Boffetta P, Rosenberg PS, Berndt SI, Karami S,
Menashe I, Yeager M, Chanock SJ, Zaridze D, Matteev V,
Janout V, Kollarova H, Bencko V, Navratilova M, Szeszenia-
Dabrowska N, Mates D, Rothman N, Brennan P, Chow WH,
Moore LE (2010) Variants in blood pressure genes and the risk
of renal cell carcinoma. Carcinogenesis 31(4):614–620. https ://
doi.org/10.1093/carci n/bgp32 1
55. Pringle KG, Delforce SJ, Wang Y, Ashton KA, Proietto A, Otton
G, Blackwell CC, Scott RJ, Lumbers ER (2016) Renin-angioten-
sin system gene polymorphisms and endometrial cancer. Endocr
Connect 5(3):128–135. https ://doi.org/10.1530/EC-15-0112
56. Deckers IA, van den Brandt PA, van Engeland M, van Schooten
FJ, Godschalk RW, Keszei AP, Schouten LJ (2015) Polymor-
phisms in genes of the renin-angiotensin-aldosterone system and
renal cell cancer risk: interplay with hypertension and intakes of
sodium, potassium and fluid. Int J Cancer 136(5):1104–1116.
https ://doi.org/10.1002/ijc.29060
Journal of Neuro-Oncology
1 3
57. Tamarat R, Silvestre JS, Durie M, Levy BI (2002) Angioten-
sin II angiogenic effect invivo involves vascular endothelial
growth factor- and inflammation-related pathways. Lab Invest
82(6):747–756
58. Wolf G, Wenzel U, Burns KD, Harris RC, Stahl RA, Thaiss
F (2002) Angiotensin II activates nuclear transcription factor-
kappaB through AT1 and AT2 receptors. Kidney Int 61(6):1986–
1995. https ://doi.org/10.1046/j.1523-1755.2002.00365 .x
59. Leung PS, Suen PM, Ip SP, Yip CK, Chen G, Lai PB (2003)
Expression and localization of AT1 receptors in hepatic Kupffer
cells: its potential role in regulating a fibrogenic response. Regul
Pept 116(1–3):61–69
60. Arrieta O, Villarreal-Garza C, Vizcaino G, Pineda B, Hernandez-
Pedro N, Guevara-Salazar P, Wegman-Ostrosky T, Villanueva-
Rodriguez G, Gamboa-Dominguez A (2015) Association
between AT1 and AT2 angiotensin II receptor expression with
cell proliferation and angiogenesis in operable breast cancer.
Tumour Biol 36(7):5627–5634. https ://doi.org/10.1007/s1327
7-015-3235-3
61. Kawai T, Forrester SJ, O’Brien S, Baggett A, Rizzo V, Egu-
chi S (2017) AT1 receptor signaling pathways in the cardio-
vascular system. Pharmacol Res 125(Pt A):4–13. https ://doi.
org/10.1016/j.phrs.2017.05.008
62. Lanz TV, Ding Z, Ho PP, Luo J, Agrawal AN, Srinagesh H,
Axtell R, Zhang H, Platten M, Wyss-Coray T, Steinman L (2010)
Angiotensin II sustains brain inflammation in mice via TGF-beta.
J Clin Invest 120(8):2782–2794. https ://doi.org/10.1172/JCI41
709
63. Levin VA, Chan J, Datta M, Yee JL, Jain RK (2017) Effect of
angiotensin system inhibitors on survival in newly diagnosed
glioma patients and recurrent glioblastoma patients receiving
chemotherapy and/or bevacizumab. J Neuro Oncol 134(2):325–
330. https ://doi.org/10.1007/s1106 0-017-2528-3
64. Clere N, Corre I, Faure S, Guihot AL, Vessieres E, Chalopin M,
Morel A, Coqueret O, Hein L, Delneste Y, Paris F, Henrion D
(2010) Deficiency or blockade of angiotensin II type 2 receptor
delays tumorigenesis by inhibiting malignant cell proliferation
and angiogenesis. Int J Cancer 127(10):2279–2291. https ://doi.
org/10.1002/ijc.25234
65. Yoshida T, Kinoshita H, Fukui K, Matsuzaki T, Yoshida K,
Mishima T, Yanishi M, Komai Y, Sugi M, Inoue T, Murota T,
Matsuda T (2017) Prognostic impact of renin-angiotensin inhibi-
tors in patients with bladder cancer undergoing radical cystec-
tomy. Ann Surg Oncol 24(3):823–831. https ://doi.org/10.1245/
s1043 4-016-5534-3
66. Moreno-Munoz D, de la Haba-Rodriguez JR, Conde F, Lopez-
Sanchez LM, Valverde A, Hernandez V, Martinez A, Villar
C, Gomez-Espana A, Porras I, Rodriguez-Ariza A, Aranda E
(2015) Genetic variants in the renin-angiotensin system predict
response to bevacizumab in cancer patients. Eur J Clin Invest
45(12):1325–1332. https ://doi.org/10.1111/eci.12557
67. Passos-Silva DG, Brandan E, Santos RA (2015) Angiotensins as
therapeutic targets beyond heart disease. Trends Pharmacol Sci
36(5):310–320. https ://doi.org/10.1016/j.tips.2015.03.001
68. Weller M, Pfister SM, Wick W, Hegi ME, Reifenberger G, Stupp
R (2013) Molecular neuro-oncology in clinical practice: a new
horizon. Lancet Oncol 14(9):e370–e379. https ://doi.org/10.1016/
s1470 -2045(13)70168 -2
69. Thakkar JP, Dolecek TA, Horbinski C, Ostrom QT, Lightner
DD, Barnholtz-Sloan JS, Villano JL (2014) Epidemiologic and
molecular prognostic review of glioblastoma. Cancer Epide-
miol 23(10):1985–1996. https ://doi.org/10.1158/1055-9965.
EPI-14-0275
70. Bradshaw AR, Wickremesekera AC, Brasch HD, Chib-
nall AM, Davis PF, Tan ST, Itinteang T (2016) Glioblas-
toma multiforme cancer stem cells express components of
the renin-angiotensin system. Front Surg 3:51. https ://doi.
org/10.3389/fsurg .2016.00051
71. Urup T, Michaelsen SR, Olsen LR, Toft A, Christensen IJ, Grun-
net K, Winther O, Broholm H, Kosteljanetz M, Issazadeh-Navi-
kas S, Poulsen HS, Lassen U (2016) Angiotensinogen and HLA
class II predict bevacizumab response in recurrent glioblastoma
patients. Mol Oncol 10(8):1160–1168. https ://doi.org/10.1016/j.
molon c.2016.05.005
72. Perdomo-Pantoja A, Mejia-Perez S, Gomez-Amador J, Wegman-
Ostrosky T (2017) Serum rs5050 AGT polymorphism is related
to poor prognosis in astrocytoma: potential biomarker in blood.
Turk Neurosurg 27(5 Suppl):102–103
73. Wegman-Ostrosky T (2014) Identificación de variantes en el gen
AGT en pacientes con astrocitoma (tesis de doctorado). Univer-
sidad de Guadalajara, Mexico
74. Medeiros R, Vasconcelos A, Costa S, Pinto D, Lobo F, Morais A,
Oliveira J, Lopes C (2004) Linkage of angiotensin I-converting
enzyme gene insertion/deletion polymorphism to the progression
of human prostate cancer. J Pathol 202(3):330–335. https ://doi.
org/10.1002/path.1529
75. Sierra Diaz E, Sanchez Corona J, Rosales Gomez RC, Gutier-
rez Rubio SA, Vazquez Camacho JG, Solano Moreno H, Moran
Moguel MC (2009) Angiotensin-converting enzyme insertion/
deletion and angiotensin type 1 receptor A1166C polymorphisms
as genetic risk factors in benign prostatic hyperplasia and pros-
tate cancer. J Renin-Angiotensin-Aldosterone Syst 10(4):241–
246. https ://doi.org/10.1177/14703 20309 35280 0
76. Devic Pavlic S, Ristic S, Flego V, Kapovic M, Radojcic Badovi-
nac A (2012) Angiotensin-converting enzyme insertion/deletion
gene polymorphism in lung cancer patients. Genet Test Mol Bio-
mark 16(7):722–725. https ://doi.org/10.1089/gtmb.2011.0306
77. Lukic S, Nikolic A, Alempijevic T, Popovic D, Sokic Miluti-
novic A, Ugljesic M, Knezevic S, Milicic B, Dinic D, Rado-
jkovic D (2011) Angiotensin-converting enzyme gene insertion/
deletion polymorphism in patients with chronic pancreatitis
and pancreatic cancer. Dig Surg 28(4):258–262. https ://doi.
org/10.1159/00032 8666
78. Srivastava K, Srivastava A, Mittal B (2010) Angiotensin I-con-
verting enzyme insertion/deletion polymorphism and increased
risk of gall bladder cancer in women. DNA Cell Biol 29(8):417–
422. https ://doi.org/10.1089/dna.2010.1033
79. Alves Correa SA, Ribeiro de Noronha SM, Nogueira-de-Souza
NC, Valleta de Carvalho C, Massad Costa AM, Juvenal Linhares
J, Vieira Gomes MT, Guerreiro da Silva ID (2009) Association
between the angiotensin-converting enzyme (insertion/deletion)
and angiotensin II type 1 receptor (A1166C) polymorphisms and
breast cancer among Brazilian women. J Renin-Angiotensin-
Aldosterone Syst 10 (1):51–58. https ://doi.org/10.1177/14703
20309 10231 7
80. Vairaktaris E, Yapijakis C, Tsigris C, Vassiliou S, Derka S,
Nkenke E, Spyridonidou S, Vylliotis A, Vorris E, Ragos V,
Neukam FW, Patsouris E (2007) Association of angiotensin-
converting enzyme gene insertion/deletion polymorphism with
increased risk for oral cancer. Acta Oncol 46(8):1097–1102. https
://doi.org/10.1080/02841 86070 13735 79
81. Rigat B, Hubert C, Alhenc-Gelas F, Cambien F, Corvol P, Sou-
brier F (1990) An insertion/deletion polymorphism in the angio-
tensin I-converting enzyme gene accounting for half the variance
of serum enzyme levels. J Clin Invest 86(4):1343–1346. https ://
doi.org/10.1172/JCI11 4844
82. Tiret L, Rigat B, Visvikis S, Breda C, Corvol P, Cambien F,
Soubrier F (1992) Evidence, from combined segregation and
linkage analysis, that a variant of the angiotensin I-converting
enzyme (ACE) gene controls plasma ACE levels. Am J Hum
Genet 51(1):197–205
Journal of Neuro-Oncology
1 3
83. Lian M, Jiang H, Wang H, Guo S (2015) Angiotensin-converting
enzyme insertion/deletion gene polymorphisms is associated with
risk of glioma in a Chinese population. J Renin-Angiotensin-
Aldosterone Syst 16(2):443–447. https ://doi.org/10.1177/14703
20313 49591 0
84. Azevedo H, Fujita A, Bando SY, Iamashita P, Moreira-Filho
CA (2014) Transcriptional network analysis reveals that AT1
and AT2 angiotensin II receptors are both involved in the reg-
ulation of genes essential for glioma progression. PLoS ONE
9(11):e110934. https ://doi.org/10.1371/journ al.pone.01109 34
85. Herrera M, Sparks MA, Alfonso-Pecchio AR, Harrison-Bernard
LM, Coffman TM (2013) Response to lack of specificity of
commercial antibodies leads to misidentification of angiotensin
type-1 receptor protein. Hypertension 61(4):e32
86. Ni L, Feng Y, Wan H, Ma Q, Fan L, Qian Y, Li Q, Xiang Y, Gao
B (2012) Angiotensin-(1–7) inhibits the migration and invasion
of A549 human lung adenocarcinoma cells through inactivation
of the PI3K/Akt and MAPK signaling pathways. Oncol Rep
27(3):783–790. https ://doi.org/10.3892/or.2011.1554
87. Liu B, Liu Y, Jiang Y (2015) Podocalyxin promotes glioblastoma
multiforme cell invasion and proliferation by inhibiting angio-
tensin-(1–7)/Mas signaling. Oncol Rep 33(5):2583–2591. https
://doi.org/10.3892/or.2015.3813
88. Ma TK, Kam KK, Yan BP, Lam YY (2010) Renin-angiotensin-
aldosterone system blockade for cardiovascular diseases: current
status. Br J Pharmacol 160(6):1273–1292. https ://doi.org/10.11
11/j.1476-5381.2010.00750 .x
89. Ronquist G, Rodriguez LA, Ruigomez A, Johansson S, Wallander
MA, Frithz G, Svardsudd K (2004) Association between capto-
pril, other antihypertensive drugs and risk of prostate cancer.
Prostate 58(1):50–56. https ://doi.org/10.1002/pros.10294
90. Friis S, Sorensen HT, Mellemkjaer L, McLaughlin JK, Nielsen
GL, Blot WJ, Olsen JH (2001) Angiotensin-converting enzyme
inhibitors and the risk of cancer: a population-based cohort study
in Denmark. Cancer 92(9):2462–2470
91. Meier CR, Derby LE, Jick SS, Jick H (2000) Angiotensin-con-
verting enzyme inhibitors, calcium channel blockers, and breast
cancer. Arch Intern Med 160(3):349–353
92. Lund EL, Spang-Thomsen M, Skovgaard-Poulsen H, Kristjansen
PE (1998) Tumor angiogenesis–a new therapeutic target in glio-
mas. Acta Neurol Scand 97(1):52–62
93. Wang D, Huang HJ, Kazlauskas A, Cavenee WK (1999) Induc-
tion of vascular endothelial growth factor expression in endothe-
lial cells by platelet-derived growth factor through the activation
of phosphatidylinositol 3-kinase. Cancer Res 59(7):1464–1472
94. Juillerat-Jeanneret L, Lohm S, Hamou MF, Pinet F (2000) Regu-
lation of aminopeptidase A in human brain tumor vasculature:
evidence for a role of transforming growth factor-beta. Lab Invest
80(6):973–980
95. Kakinuma Y, Hama H, Sugiyama F, Yagami K, Goto K,
Murakami K, Fukamizu A (1998) Impaired blood-brain barrier
function in angiotensinogen-deficient mice. Nat Med 4(9):1078–
1080. https ://doi.org/10.1038/2070
96. Egidy G, Eberl LP, Valdenaire O, Irmler M, Majdi R, Diserens
AC, Fontana A, Janzer RC, Pinet F, Juillerat-Jeanneret L (2000)
The endothelin system in human glioblastoma. Lab Invest
80(11):1681–1689
97. Arrieta O, Guevara P, Escobar E, Garcia-Navarrete R, Pineda
B, Sotelo J (2005) Blockage of angiotensin II type I receptor
decreases the synthesis of growth factors and induces apoptosis
in C6 cultured cells and C6 rat glioma. Br J Cancer 92(7):1247–
1252. https ://doi.org/10.1038/sj.bjc.66024 83
98. Stoll M, Steckelings UM, Paul M, Bottari SP, Metzger R, Unger
T (1995) The angiotensin AT2-receptor mediates inhibition of
cell proliferation in coronary endothelial cells. J Clin Invest
95(2):651–657. https ://doi.org/10.1172/JCI11 7710
99. Fogarty DJ, Sanchez-Gomez MV, Matute C (2002) Multiple
angiotensin receptor subtypes in normal and tumor astrocytes
invitro. Glia 39(3):304–313. https ://doi.org/10.1002/glia.10117
100. Januel E, Ursu R, Alkhafaji A, Marantidou A, Doridam J, Belin
C, Levy-Piedbois C, Carpentier AF (2015) Impact of renin-angi-
otensin system blockade on clinical outcome in glioblastoma. Eur
J Neurol 22(9):1304–1309. https ://doi.org/10.1111/ene.12746
101. Carpentier AF, Ferrari D, Bailon O, Ursu R, Banissi C,
Dubessy AL, Belin C, Levy C (2012) Steroid-sparing
effects of angiotensin-II inhibitors in glioblastoma patients.
Eur J Neurol 19(10):1337–1342. https ://doi.org/10.111
1/j.1468-1331.2012.03766 .x
102. Kourilsky A, Bertrand G, Ursu R, Doridam J, Barlog C, Faillot T,
Mandonnet E, Belin C, Levy C, Carpentier AF (2016) Impact of
angiotensin-II receptor blockers on vasogenic edema in glioblas-
toma patients. J Neurol 263(3):524–530. https ://doi.org/10.1007/
s0041 5-015-8016-9
103. Saavedra JM (2012) Angiotensin II AT(1) receptor block-
ers as treatments for inflammatory brain disorders. Clin Sci
123(10):567–590. https ://doi.org/10.1042/CS201 20078
104. Saavedra JM (2017) Beneficial effects of angiotensin II receptor
blockers in brain disorders. Pharmacol Res 125(Pt A):91–103.
https ://doi.org/10.1016/j.phrs.2017.06.017
105. Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T (2010)
Angiotensin II induced cerebral microvascular inflammation and
increased blood-brain barrier permeability via oxidative stress.
Neuroscience 171(3):852–858. https ://doi.org/10.1016/j.neuro
scien ce.2010.09.029
106. Fleegal-DeMotta MA, Doghu S, Banks WA (2009) Angioten-
sin II modulates BBB permeability via activation of the AT(1)
receptor in brain endothelial cells. J Cereb Blood Flow Metab
29(3):640–647. https ://doi.org/10.1038/jcbfm .2008.158
107. Sano H, Hosokawa K, Kidoya H, Takakura N (2006) Negative
regulation of VEGF-induced vascular leakage by blockade of
angiotensin II type 1 receptor. Arterioscler Thromb Vasc Biol
26(12):2673–2680. https ://doi.org/10.1161/01.ATV.00002 45821
.77155 .c3
108. Lu-Emerson C, Duda DG, Emblem KE, Taylor JW, Gerstner ER,
Loeffler JS, Batchelor TT, Jain RK (2015) Lessons from anti-
vascular endothelial growth factor and anti-vascular endothe-
lial growth factor receptor trials in patients with glioblastoma.
J Clin Oncol 33(10):1197–1213. https ://doi.org/10.1200/
JCO.2014.55.9575
109. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishi-
kawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D,
Brandes AA, Hilton M, Abrey L, Cloughesy T (2014) Beva-
cizumab plus radiotherapy-temozolomide for newly diagnosed
glioblastoma. N Engl J Med 370(8):709–722. https ://doi.
org/10.1056/NEJMo a1308 345
110. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal
DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M,
Jeraj R, Brown PD, Jaeckle KA, Schiff D, Stieber VW, Brachman
DG, Werner-Wasik M, Tremont-Lukats IW, Sulman EP, Aldape
KD, Curran WJ Jr, Mehta MP (2014) A randomized trial of
bevacizumab for newly diagnosed glioblastoma. N Engl J Med
370(8):699–708. https ://doi.org/10.1056/NEJMo a1308 573
111. Levin VA, Mendelssohn ND, Chan J, Stovall MC, Peak SJ,
Yee JL, Hui RL, Chen DM (2015) Impact of bevacizumab
administered dose on overall survival of patients with progres-
sive glioblastoma. J Neuro Oncol 122(1):145–150. https ://doi.
org/10.1007/s1106 0-014-1693-x
112. Lombardi G, Zustovich F, Farina P, Fiduccia P, Della Puppa
A, Polo V, Bertorelle R, Gardiman MP, Banzato A, Ciccarino
P, Denaro L, Zagonel V (2013) Hypertension as a biomarker
in patients with recurrent glioblastoma treated with antiangio-
genic drugs: a single-center experience and a critical review
Journal of Neuro-Oncology
1 3
of the literature. Anticancer Drugs 24(1):90–97. https ://doi.
org/10.1097/CAD.0b013 e3283 5aa5f d
113. Ranpura V, Pulipati B, Chu D, Zhu X, Wu S (2010) Increased
risk of high-grade hypertension with bevacizumab in cancer
patients: a meta-analysis. Am J Hypertens 23(5):460–468. https
://doi.org/10.1038/ajh.2010.25
114. Emile G, Pujade-Lauraine E, Alexandre J (2014) Should we use
the angiotensin-converting enzyme inhibitors for the treatment of
anti-VEGF-induced hypertension? Ann Oncol 25(8):1669–1670.
https ://doi.org/10.1093/annon c/mdu19 7
115. Okwan-Duodu D, Landry J, Shen XZ, Diaz R (2013) Angioten-
sin-converting enzyme and the tumor microenvironment: mecha-
nisms beyond angiogenesis. Am J Physiol Regul Integr Comp
Physiol 305(3):R205-215. https ://doi.org/10.1152/ajpre gu.00544
.2012
116. Tom B, Dendorfer A, de Vries R, Saxena PR, Jan Danser AH
(2002) Bradykinin potentiation by ACE inhibitors: a matter
of metabolism. Br J Pharmacol 137(2):276–284. https ://doi.
org/10.1038/sj.bjp.07048 62
117. Stylianopoulos T, Jain RK (2013) Combining two strategies to
improve perfusion and drug delivery in solid tumors. Proc Natl
Acad Sci USA 110(46):18632–18637. https ://doi.org/10.1073/
pnas.13184 15110
118. Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin
SV, Stylianopoulos T, Mousa AS, Han X, Adstamongkonkul P,
Popovic Z, Huang P, Bawendi MG, Boucher Y, Jain RK (2013)
Angiotensin inhibition enhances drug delivery and potentiates
chemotherapy by decompressing tumour blood vessels. Nat
Commun 4:2516. https ://doi.org/10.1038/ncomm s3516
119. Harrison LB, Chadha M, Hill RJ, Hu K, Shasha D (2002) Impact
of tumor hypoxia and anemia on radiation therapy outcomes.
Oncologist 7(6):492–508
120. Clarke RH, Moosa S, Anzivino M, Wang Y, Floyd DH, Purow
BW, Lee KS (2014) Sustained radiosensitization of hypoxic
glioma cells after oxygen pretreatment in an animal model of
glioblastoma and invitro models of tumor hypoxia. PLoS ONE
9(10):e111199. https ://doi.org/10.1371/journ al.pone.01111 99
121. Pinter M, Jain RK (2017) Targeting the renin-angiotensin system
to improve cancer treatment: implications for immunotherapy.
Sci Transl Med. https ://doi.org/10.1126/scitr anslm ed.aan56 16
122. Liu H, Naxerova K, Pinter M, Incio J, Lee H, Shigeta K, Ho WW,
Crain JA, Jacobson A, Michelakos T, Dias-Santos D, Zanconato
A, Hong TS, Clark JW, Murphy JE, Ryan DP, Deshpande V, Lil-
lemoe KD, Fernandez-Del Castillo C, Downes M, Evans RM,
Michaelson J, Ferrone CR, Boucher Y, Jain RK (2017) Use of
angiotensin system inhibitors is associated with immune acti-
vation and longer survival in nonmetastatic pancreatic ductal
adenocarcinoma. Clin Cancer Res 23(19):5959–5969. https ://doi.
org/10.1158/1078-0432.CCR-17-0256
123. Pinter M, Weinmann A, Worns MA, Hucke F, Bota S, Marquardt
JU, Duda DG, Jain RK, Galle PR, Trauner M, Peck-Radosavljevic
M, Sieghart W (2017) Use of inhibitors of the renin-angiotensin
system is associated with longer survival in patients with hepa-
tocellular carcinoma. United Eur Gastroenterol J 5(7):987–996.
https ://doi.org/10.1177/20506 40617 69569 8
124. Rosen EM, Day R, Singh VK (2014) New approaches to radia-
tion protection. Front Oncol 4:381. https ://doi.org/10.3389/
fonc.2014.00381
125. Johnke RM, Sattler JA, Allison RR (2014) Radioprotective agents
for radiation therapy: future trends. Future Oncol 10(15):2345–
2357. https ://doi.org/10.2217/fon.14.175
126. McLaughlin MF, Donoviel DB, Jones JA (2017) Novel indica-
tions for commonly used medications as radiation protectants in
spaceflight. Aerosp Med Hum Perform 88(7):665–676. https ://
doi.org/10.3357/AMHP.4735.2017
127. Robbins ME, Payne V, Tommasi E, Diz DI, Hsu FC, Brown WR,
Wheeler KT, Olson J, Zhao W (2009) The AT1 receptor antag-
onist, L-158,809, prevents or ameliorates fractionated whole-
brain irradiation-induced cognitive impairment. Int J Radiat
Oncol Biol Phys 73(2):499–505. https ://doi.org/10.1016/j.ijrob
p.2008.09.058
128. Lee TC, Greene-Schloesser D, Payne V, Diz DI, Hsu FC,
Kooshki M, Mustafa R, Riddle DR, Zhao W, Chan MD, Rob-
bins ME (2012) Chronic administration of the angiotensin-con-
verting enzyme inhibitor, ramipril, prevents fractionated whole-
brain irradiation-induced perirhinal cortex-dependent cognitive
impairment. Radiation Res 178(1):46–56
129. Jenrow KA, Brown SL, Liu J, Kolozsvary A, Lapanowski K, Kim
JH (2010) Ramipril mitigates radiation-induced impairment of
neurogenesis in the rat dentate gyrus. Radiat Oncol 5:6. https ://
doi.org/10.1186/1748-717X-5-6
130. Kast RE, Boockvar JA, Bruning A, Cappello F, Chang WW, Cvek
B, Dou QP, Duenas-Gonzalez A, Efferth T, Focosi D, Ghaffari
SH, Karpel-Massler G, Ketola K, Khoshnevisan A, Keizman D,
Magne N, Marosi C, McDonald K, Munoz M, Paranjpe A, Pourg-
holami MH, Sardi I, Sella A, Srivenugopal KS, Tuccori M, Wang
W, Wirtz CR, Halatsch ME (2013) A conceptually new treatment
approach for relapsed glioblastoma: coordinated undermining of
survival paths with nine repurposed drugs (CUSP9) by the inter-
national initiative for accelerated improvement of glioblastoma
care. Oncotarget 4(4):502–530. https ://doi.org/10.18632 /oncot
arget .969
131. Kast RE, Karpel-Massler G, Halatsch ME (2014) CUSP9* treat-
ment protocol for recurrent glioblastoma: aprepitant, artesunate,
auranofin, captopril, celecoxib, disulfiram, itraconazole, ritona-
vir, sertraline augmenting continuous low dose temozolomide.
Oncotarget 5(18):8052–8082. https ://doi.org/10.18632 /oncot
arget .2408
132. Assistance Publique – Hôpitaux de Paris. Angiotensin II
Receptor Blockers, Steroids and Radiotherapy in Glioblastoma
(ASTER). https ://clini caltr ials.gov/ct2/show/NCT01 80545 3
(Accessed 16 Jan 2018)
... On the contrary, binding of Ang II to AT2R inhibits angiogenesis, cell proliferation, migration, invasion, and metastasis. Binding of Ang-(1-7) with MASR exerts similar anti-tumor effects clear cell carcinoma [12], and gliomas [13], etc. The importance of RAS and its components in cancer development merits attentions to cancer researchers. ...
... Tumor angiogenesis produced by RAS is mainly induced by vascular endothelial growth factor (VEGF) and its related pathways. As opposed to the systemic RAS, recently, the concept of a localized RAS has been reported, such as in the central nervous system (CNS) [13] and female reproductive organs [25] (Fig. 16.3). ...
Chapter
Full-text available
Renin-Angiotensin System (RAS) is a vital system regulating blood pressure and maintaining sodium homeostasis in the human body. It consists of Angiotensin I (Ang I), Angiotensin II (Ang II), Angiotensin-converting enzyme (ACE), Angiotensin II type 1 receptor (AT1R), and angiotensin II type 2 receptor (AT2R), which functions in both normal and pathological conditions including cancer. Besides, the effectors of RAS are also included, such as Angiotensin-(1-7). This review focuses on the pre-clinical studies and clinical trials assessing the roles of RAS in regulating tumor progression as well as the underlying mechanisms.KeywordsRenin-angiotensin systemCancer progressionProliferationAngiogenesisMetastasis
... AGT codes for angiotensinogen, an element of the reninangiotensin system (RAS). RAS elements have been found in astrocytoma tissue and have been associated with prognosis [8]. High AGT expression levels and low methylation levels of its promoter are predictive of a poor response to bevacizumab in recurrent GB [9,10]. ...
... Low AGT levels and AGT epigenetic silencing have been associated with a positive bevacizumab response in patients with recurrent GB [9,10]. In observational studies, it has been reported that patients receiving RAS inhibitors had longer OS [8]. Nonetheless, in the only randomized clinical trial published thus far, the use of an angiotensin II receptor blocker was not associated with improved survival [31]. ...
Article
Full-text available
Purpose Astrocytomas are a type of malignant brain tumor with an unfavorable clinical course. The impact of AGT and MGMT somatic variants in the prognosis of astrocytoma is unknown, and it is controversial for TP53. Moreover, there is a lack of knowledge regarding the molecular characteristics of astrocytomas in Mexican patients. Methods We studied 48 Mexican patients, men and women, with astrocytoma (discovery cohort). We performed DNA deep sequencing in tumor samples, targeting AGT, MGMT and TP53, and we studied MGMT gene promoter methylation status. Then we compared our findings to a cohort which included data from patients with astrocytoma from The Cancer Genome Atlas (validation cohort). Results In the discovery cohort, we found a higher number of somatic variants in AGT and MGMT than in the validation cohort (10.4% vs < 1%, p < 0.001), and, in both cohorts, we observed only women carried variants AGT variants. We also found that the presence of either MGMT variant or promoter methylation was associated to better survival and response to chemotherapy, and, in conjunction with TP53 variants, to progression-free survival. Conclusions The occurrence of AGT variants only in women expands our knowledge about the molecular differences in astrocytoma between men and women. The increased prevalence of AGT and MGMT variants in the discovery cohort also points towards possible distinctions in the molecular landscape of astrocytoma among populations. Our findings warrant further study.
... These receptors are proposed to have opposing actions, whereby AT 1 R promotes cell growth and angiogenesis and AT 2 R counteracts these effects by functioning as an endogenous antagonist of the AT 1 R (2). The dysregulation of RAS through the aberrant expression of its components has been reported in many pathophysiological conditions, including cancer, where overexpression of AT 1 R has been correlated with more aggressive disease (3)(4)(5). Thus, AT 1 R antagonists such as the sartans have been proposed as agents for systemic anticancer therapy (6). ...
Article
Full-text available
Glioblastoma (GBM) is an aggressive malignant primary brain tumor with limited therapeutic options. We show that the angiotensin II (AngII) type 2 receptor (AT 2 R) is a therapeutic target for GBM and that AngII, endogenously produced in GBM cells, promotes proliferation through AT 2 R. We repurposed EMA401, an AT 2 R antagonist originally developed as a peripherally restricted analgesic, for GBM and showed that it inhibits the proliferation of AT 2 R-expressing GBM spheroids and blocks their invasiveness and angiogenic capacity. The crystal structure of AT 2 R bound to EMA401 was determined and revealed the receptor to be in an active-like conformation with helix-VIII blocking G-protein or β-arrestin recruitment. The architecture and interactions of EMA401 in AT 2 R differ drastically from complexes of AT 2 R with other relevant compounds. To enhance central nervous system (CNS) penetration of EMA401, we exploited the crystal structure to design an angiopep-2–tethered EMA401 derivative, A3E. A3E exhibited enhanced CNS penetration, leading to reduced tumor volume, inhibition of proliferation, and increased levels of apoptosis in an orthotopic xenograft model of GBM.
... The studies that focused on RAS components in CRC generally found a consistent correlation between RAS-related gene expression in CRC tissues with ACE, MasR, AT1R, and AT2R expression being altered in CRC primary and metastatic tissues [15,27]. Protumoral associations of the RAS proteins may be related to gliomas as well [26]. ...
Article
Full-text available
Materials and methods: Quantitative expression of the RNA of these 17 genes in normal and cancerous tissues obtained using chip arrays from the public functional genomics data repository, Gene Expression Omnibus (GEO) application, was compared statistically. Results: Expression of four genes, AGT (angiotensinogen), ENPEP (aminopeptidase A) MME (neprilysin), and PREP (prolyl endopeptidase), was significantly upregulated in CRC specimens. Expression of REN (renin), THOP (thimet oligopeptidase), NLN (neurolysin), PRCP (prolyl carboxypeptidase), ANPEP (aminopeptidase N), and MAS1 (Mas receptor) was downregulated in CRC specimens. Conclusions: Presuming gene expression parallel protein expression, these results suggest that increased production of the angiotensinogen precursor of angiotensin (ANG) peptides, with the reduction of the enzymes that metabolize it to ANG II, can lead to accumulation of angiotensinogen in CRC tissues. Downregulation of THOP, NLN, PRCP, and MAS1 gene expression, whose proteins contribute to the ACE2/ANG 1-7/Mas axis, suggests that reduced activity of this RAS branch could be permissive for oncogenicity. Components of the RAS may be potential therapeutic targets for treatment of CRC.
... solid tumors, such as in the breast, liver, and gastrointestinal tract tumors (Figure 1), and they are involved in the pathological and physiological processes of cancer, including proliferation, migration, apoptosis, and angiogenesis, suggesting that dysfunction of RAS contributes to tumor progression [4,[14][15][16][17]. At the same time, abnormal expression levels of RAS have ing cytotoxicity and inducing cell apoptosis [24]. ...
Article
The renin-angiotensin system (RAS) regulates physiological functions of the cardiovascular system, kidneys, and other tissues. Various in vivo and in vitro studies have shown that RAS plays a pivotal role in the development of malignant tumors, while several retrospective studies have confirmed that patients undergoing long-term RAS inhibitors (RASi) treatment have a lowered risk of cancer. Moreover, blocking RAS has been shown to inhibit tumor growth, metastasis, and angiogenesis in various experimental models of malignant tumors. Herein, we review the available RASi-related literature and provide an analysis using the scientific atlas software VOSviewer. We observed that recent studies have primarily focused on gene expression, tumor biology, and survival analysis. Through an in-depth data analysis from the Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx), we identified the impact of AGTR1, an essential component of RAS, on tumors, and we discuss the underlying biological mechanism of RASi. Furthermore, we outline the research progress and potential use of RASi in tumor treatment. Overall, RASi may be a promising adjunct in cancer therapy.
Chapter
The renin angiotensin-system, its well-known systemic regulation of the systemic pressure and its role in hypertension has been widely studies, but this system has also a paracrine function. The RAS was been found to be express in almost every tissue, and there is enough evidence to link it to carcinogenesis and cancer progression. With the purpose of facilitating advances in the study of this disease, Weinberg and Hanahan described a series of cellular characteristics that tumor cells can share, which are known as the Hallmarks of cancer. These are defined as “the distinctive and complementary capacities that cells must acquire during the carcinogenesis process that allow them to survive, proliferate and disseminate”. Originally there were 6, but currently up to 10 are “officially” accepted and 4 new ones were proposed by Hanahan in early 2022. In this chapter we describe how RAS is related to the 10 classical hallmarks of cancer, defined as a series of cellular characteristics that tumor cells can share, but also to the 4 new hallmarks described recently.KeywordsRenin-angiotensinCancerHallmarksAngiotensinogenAngiotensinACE2ACEEpigeneticsGenomics
Article
Background: Captopril is a well-characterized, FDA-approved drug that has demonstrated promise as a repurposed oncology therapeutic. Captopril's known anti-cancer effects include inhibition of Matrix Metalloproteinase-2 (MMP-2), an endopeptidase which selectively breaks down the extracellular matrix to promote cell migration. MMP-2 is a known therapeutic target in gliomas, tumors with significant clinical need. Using an aggressive gliosarcoma model, we assessed captopril's effects on MMP-2 expression in vitro and in vivo as well as its efficacy as an adjuvant in combination therapy regimens in vivo. Methods: Following captopril treatment, MMP-2 protein expression and migratory capabilities of 9 L gliosarcoma cells were assessed in vitro via western blots and scratch wound assays, respectively. Rats were intracranially implanted with 9 L gliosarcoma tumors, and survival was assessed in the following groups: control; captopril (30 mg/kg/day); temozolomide (TMZ) (50 mg/kg/day), and captopril+TMZ. In vivo experiments were accompanied by immunohistochemistry for MMP-2 from brain tissue. Results: In vitro, captopril decreased MMP-2 protein expression and reduced migratory capacity in 9 L gliosarcoma cells. In a gliosarcoma animal model, captopril decreased MMP-2 protein expression and extended survival as a TMZ adjuvant relative to untreated controls, captopril monotherapy, and TMZ monotherapy groups (27.5 versus 14 (p < 0.001), 16 (p < 0.001), and 23 (p = 0.018) days, respectively). Conclusions: Captopril decreases gliosarcoma cell migration, which may be mediated by reduction in MMP-2 protein expression. Captopril provided a survival advantage as a TMZ adjuvant in a rat intracranial gliosarcoma model. Captopril may represent a promising potential adjuvant to TMZ therapy in gliosarcoma as a modulator of the MMP-2 pathway.
Article
Full-text available
Background: History has taught us that Mexican culture has been largely supported by women, despite gender prejudice from the society. Neurosurgery has not been the exception. Therefore, we investigated the challenges and influence of female neurosurgeons in Mexico. Methods: We conducted a review of the literature and an analysis of the internal database of the Mexican Society of Neurological Surgery focusing on 3 topics: 1) the historical presence of women and gender inequality in Mexico; 2) the life and legacy of the woman who became the first neurosurgeon in Mexico and in Latin America; and 3) the participation of women in neurosurgery in the past 3 decades. Results: In Latin America, the first woman in neurosurgery was María Cristina García-Sancho, who completed her neurosurgical training in 1951. Currently, women represent 6.2% of the total members of the Mexican Society of Neurological Surgery (MSNS). This percentage is still low, although data collected in this study suggest that it might increase in the next few years because 16.7% of Board Directors of the MSNS are women, the next elected president is a female neurosurgeon, and 14.5% of neurosurgery residents are women. Conclusions: Although a steady increase has occurred of women in neurosurgery in Mexico, there is still work to do, especially to overcome the barriers related to the old assumptions of the cultural and social roles of women.
Article
Pancreatic cancer is among the most lethal malignancies with poor prognosis and patients become chemoresistant to current therapies, supporting further investigations to identify new therapeutic regimens in the treatment of this condition. Preclinical and clinical studies now appear to support the role of the renin-angiotensin system (RAS) in the regulation of tumor growth, angiogenesis, and metastasis in different malignancies including pancreatic cancer. These studies suggest that RAS blockers; Angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs); could have anti-carcinogenic effects and improve clinical outcomes in the management of pancreatic cancer. Here we provided an overview of ACE inhibitors and ARBs as a potential therapeutic option in the treatment of pancreatic cancer.
Chapter
In recent years, multiple biomarkers have emerged in the field of neuro-oncology, with useful applications in improving diagnostic accuracy, establishing patient’s prognosis and predictive response to available medical therapy. The most frequently associated biomarkers, and their respective alterations, include, IDH1/2 mutation, 1p/19q codeletion, ATRX loss, TP53 mutation, MGMT promoter methylation, BRAF and H3 mutations. In this chapter, we review these biomarkers, but also other less understood ones involved in the diagnosis, prognosis and the treatment of gliomas, such as ABC transporters, AGT mutations, G-CIMP mutation, PTEN mutation, among others. We also include two algorithms for molecular-based decision-making for pediatric and adult gliomas and tables with comprehensive information of biomarkers in gliomas.
Article
Full-text available
Renin-angiotensin system (RAS) inhibitors (RASi)—widely prescribed for the treatment of cardiovascular diseases—have considerable potential in oncology. The RAS plays a crucial role in cancer biology and affects tumor growth and dissemination directly and indirectly by remodeling the tumor microenvironment. We review clinical data on the benefit of RASi in primary and metastatic tumors and propose that, by activating immunostimulatory pathways, these inhibitors can enhance immunotherapy of cancer.
Article
Full-text available
This article reviews the histologic and molecular characterization of gliomas, including the new “integrated diagnoses” of the World Health Organization Classification, 2016 edition. The entities reviewed within include diffuse gliomas (astrocytoma, oligodendroglioma, glioblastoma), as well as circumscribed and low-grade gliomas (angiocentric glioma, pilocytic astrocytoma, subependymal giant cell astrocytoma, pleomorphic xanthoastrocytoma, pilomyxoid astrocytoma, ependymoma, myxopapillary ependymoma, and subependymoma). Diagnostic, prognostic, and predictive biomarkers are discussed for each entity. We review how molecular testing for IDH1 and ATRX and detection of chromosome 1p/19q codeletion can be used to categorize glioblastomas as IDH-wildtype or IDH-mutant, and lower grade diffuse gliomas into three molecular groups that correlate better with patient outcomes than histologic subtyping. Pediatric diffuse gliomas are highlighted, including diffuse midline glioma, H3 K27M-mutant, and inherited germline mutations that predispose to pediatric gliomas. The utility of genomic profiling of certain gliomas is discussed, including identifying candidates for experimental therapies. This review is meant to be a concise summary of glioma characterization for the practicing pathologist.
Article
Full-text available
Given prior studies that suggest the use of angiotensin system inhibitors (ASIs) is associated with prolonged overall survival (OS) in glioblastoma (GBM) patients, we evaluated the effect of ASIs in glioma patients receiving chemotherapy and/or bevacizumab (BEV). Using retrospective IRB-approved electronic chart review of newly diagnosed WHO grade 2-4 glioma patients from the Kaiser Permanente Tumor Registry of Northern California, we evaluated the impact of ASIs on OS by Cox proportional hazard model analysis for subgroups who received cytotoxic therapy, cytotoxic therapy with BEV, or BEV alone, as well as those with recurrent GBM (rGBM). Of the 1186 glioma patients who received chemotherapy ASI exposure improved OS (HR 0.82; 95% CI 0.71, 0.93; p = 0.003). When stratified by BEV exposure, a sub-analysis revealed further OS advantage for the BEV group (HR 0.75, 95% CI 0.62, 0.90; p = 0.002). In a second cohort of 181 rGBM patients who received BEV in varying dosages, ASI exposure conferred an OS advantage (HR 0.649; 95% CI 0.46, 0.92; p = 0.016). Moreover, patients with ASI exposure who received low-dose BEV treatment (AUCBEV < 3.6 mg wk/kg) had a significantly longer OS (median = 99 weeks; 95% CI 44.3, 205) than those without ASI (median OS = 55.6 weeks; 95% CI 37.7-73.7; p = 0.032). ASI use is associated with longer OS in glioma patients. Further survival advantage with ASI use was observed in rGBM patients receiving low-dose bevacizumab. These data warrant prospective evaluation of adding ASI to low-dose BEV treatment in GBM patients to improve the outcome of standard therapies.
Article
Full-text available
Purpose: Angiotensin system inhibitors (ASIs) can improve prognosis in multiple cancer types, including pancreatic ductal adenocarcinoma (PDAC). However, no study has examined the effect of ASIs alone or combined with adjuvant chemotherapy in resected PDAC patients. Experimental Design: We performed an analysis of the records of ASI users and non-user patients with PDAC seen at Massachusetts General Hospital between January 2006 and December 2010. To identify mechanisms of ASIs in PDAC, we performed RNA-Seq of resected primary lesions. Results: 794 consecutive patients were included. In 299 resected patients, ASI-users experienced longer overall survival (OS) in both univariate (median OS: 36.3 vs. 19.3 months, p=0.011) and adjusted multivariate (HR, 0.505; 95%CI, 0.339 - 0.750; p=0.001) analyses. Propensity score adjusted analysis also showed a longer median OS for chronic ASI-users. In unresected patients, the beneficial effect of ASIs was significant in patients with locally advanced disease, but not in metastatic patients. RNA-Seq analysis revealed in tumors of ASI-users (lisinopril) a normalized extracellular matrix, a reduced expression of genes involved in PDAC progression (e.g. WNT and Notch signaling) and an increased expression of genes linked with the activity of T cells and antigen-presenting cells. Finally, chronic use of ASI was associated with a gene expression signature which is predictive of survival in independent validation cohorts. Conclusions: In patients with non-metastatic PDAC, chronic ASI use is associated with longer OS independently of chemotherapy. Our RNA-Seq analysis suggests that ASI reduce the malignant potential of cancer cells and stimulate the immune microenvironment in primary PDAC.
Article
Full-text available
Background Renin–angiotensin system inhibitors (RAS inhibitors) are antihypertensive agents with potential antitumor effects. However, various studies have yielded conflicting results on the influence of RAS inhibitors on survival of cancer patients. The aim of this study was to evaluate the effect of RAS inhibitors on recurrence, metastasis, and survival in cancer patients through a meta-analysis. Methods PubMed, Web of Science, EMBASE, and Cochrane Library were systematically searched from inception to December 2016. The pooled hazard ratio (HR) with its 95% confidence interval (95% CI) was calculated to evaluate the association between RAS inhibitors and recurrence, metastasis, and survival in cancer patients. Results Fifty-five eligible studies were included in the present meta-analysis. Results showed that there were significant improvements in overall survival (OS) (HR = 0.82; 95% CI: 0.77–0.88; P < 0.001), progression-free survival (HR = 0.74; 95% CI: 0.66–0.84; P < 0.001), and disease-free survival (HR = 0.80; 95% CI: 0.67–0.95; P = 0.01) in RAS inhibitor users compared with nonusers. Subgroup analyses revealed that the effect of RAS inhibitors on OS depended on the cancer type or different RAS inhibitors. Conclusion This meta-analysis suggests that RAS inhibitors could improve the survival of cancer patients and depend on cancer type and types of RAS inhibitors.
Article
The traditional renin-angiotensin system (RAS) is indispensable system in adjusting sodium homeostasis, body fluid volume, and controlling arterial blood pressure. The key elements are renin splitting inactive angiotensinogen to yield angiotensin (Ang-I). Ang-1 is then changed by angiotensin-1 converting enzyme (ACE) into angiotensin II (Ang-II). Using PubMed, Google Scholar, and other means, we searched the peer-reviewed literature from 1990 to 2013 for articles on newly discovered findings related to the RAS, especially focusing on how the system influences the central nervous system (CNS). The classical RAS is now considered to be only part of the picture; the discovery of additional RAS pathways in the brain and elsewhere has yielded a vastly improved understanding of how the RAS influences the CNS. Newly discovered effects of the RAS on brain tissue include neuroprotection, cognition, and cerebral vasodilation. A number of brain biochemical pathways are influenced by the brain RAS. Within various pathways, there are potential opportunities for classical pharmacologic interventions as well as the possibility of controlling gene expression.
Article
A canonical brain Renin-Angiotensin System (RAS), like the canonical peripheral RAS, has long been proposed as a major regulator of brain function. Because of methodological limitations, however, this system is in urgent need of critical revision. The Angiotensin II AT1 receptors have been confirmed as key factors in the regulation of brain function, and AT1 receptor over activity has been established as a major and early injury factor in the development of many brain diseases. Consequently, Angiotensin Receptor Blockers (ARBs), compounds widely used to treat cardiovascular and metabolic disorders, are excellent candidates for repurposing for the treatment of brain disorders. This review will discuss some of the reasons why revisiting brain RAS is a pressing necessity, will present evidence for a participation of AT1 receptor over activity in the development of major brain disorders, and will present definite evidence of ARBs neuroprotective effects. The review will focus on the beneficial effects of ARB therapy in stroke, neurodegenerative disorders including Alzheimer’s and Parkinson’s diseases, traumatic brain injury, radiation- induced brain damage, stress and mood disorders.
Article
Background: In the space environment, the traditional radioprotective principles of time, distance, and shielding become difficult to implement. Additionally, the complex radiation environment inherent in space, the chronic exposure timeframe, and the presence of numerous confounding variables complicate the process of creating appropriate risk models for astronaut exposure. Pharmaceutical options hold tremendous promise to attenuate acute and late effects of radiation exposure in the astronaut population. Pharmaceuticals currently approved for other indications may also offer radiation protection, modulation, or mitigation properties along with a well-established safety profile. Currently there are only three agents which have been clinically approved to be employed for radiation exposure, and these only for very narrow indications. This review identifies a number of agents currently approved by the U.S. Food and Drug Administration (FDA) which could warrant further investigation for use in astronauts. Specifically, we examine preclinical and clinical evidence for statins, nonsteroidal anti-inflammatory drugs (NSAIDs), angiotensin converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), metformin, calcium channel blockers, β adrenergic receptor blockers, fingolimod, N-acetylcysteine, and pentoxifylline as potential radiation countermeasures.McLaughlin MF, Donoviel DB, Jones JA. Novel indications for commonly used medications as radiation protectants in spaceflight. Aerosp Med Hum Perform. 2017; 88(7):665-676.
Article
The importance of the renin angiotensin aldosterone system in cardiovascular physiology and pathophysiology has been well described whereas the detailed molecular mechanisms remain elusive. The angiotensin II type 1 receptor (AT1 receptor) is one of the key players in the renin angiotensin aldosterone system. The AT1 receptor promotes various intracellular signaling pathways resulting in hypertension, endothelial dysfunction, vascular remodeling and end organ damage. Accumulating evidence shows the complex picture of AT1 receptor-mediated signaling; AT1 receptor-mediated heterotrimeric G protein-dependent signaling, transactivation of growth factor receptors, NADPH oxidase and ROS signaling, G protein-independent signaling, including the β-arrestin signals and interaction with several AT1 receptor interacting proteins. In addition, there is functional cross-talk between the AT1 receptor signaling pathway and other signaling pathways. In this review, we will summarize an up to date overview of essential AT1 receptor signaling events and their functional significances in the cardiovascular system.
Article
Background: Inhibition of the renin-angiotensin system (RAS) was associated with longer survival in patients with different solid malignancies. Objective: The objective of this study was to investigate the effect of RAS inhibitor (RASi) treatment (angiotensin-converting enzyme inhibitors or angiotensin-II-receptor blockers) on survival of patients with hepatocellular carcinoma (HCC). Methods: Patients diagnosed with HCC and Child-Pugh A between 1992 and 2013 who received sorafenib, experimental therapy, or best supportive care were eligible for the Vienna cohort. The Mainz cohort included patients with HCC and Child-Pugh A who received sorafenib treatment between 2007 and 2016. The association between RASi and overall survival (OS) was evaluated in univariate and multivariate analyses. Results: In the Vienna cohort, 43 of 156 patients received RASi for hypertension. RASi treatment was associated with longer OS (11.9 vs. 6.8 months (mo); p = 0.014) and remained a significant prognostic factor upon multivariate analysis (HR = 0.6; 95% CI 0.4-0.9; p = 0.011). In subgroup analysis, patients treated with sorafenib plus RASi had better median OS (19.5 mo) compared to those treated with either sorafenib (10.9 mo) or RASi (9.7 mo) alone (p = 0.043). The beneficial effect of RASi on survival was confirmed in the Mainz cohort (n = 76). Conclusion: RAS inhibition is associated with longer survival in HCC patients with Child-Pugh class A.