You are on page 1of 14

REVIEW

For reprint orders, please contact: reprints@futuremedicine.com

Circulating biomarker panels for targeted


therapy in brain tumors

Cristiana Tanase*,1, Radu Albulescu1,2, Elena Codrici1, Ionela Daniela Popescu1, Simona
Mihai1, Ana Maria Enciu1,3, Maria Linda Cruceru3, Adrian Claudiu Popa3, Ana Iulia
Neagu1,4, Laura Georgiana Necula1,4, Cristina Mambet1,4 & Monica Neagu1

ABSTRACT An important goal of oncology is the development of cancer risk-identifier


biomarkers that aid early detection and target therapy. High-throughput profiling represents
a major concern for cancer research, including brain tumors. A promising approach for
efficacious monitoring of disease progression and therapy could be circulating biomarker
panels using molecular proteomic patterns. Tailoring treatment by targeting specific
protein–protein interactions and signaling networks, microRNA and cancer stem cell
signaling in accordance with tumor phenotype or patient clustering based on biomarker
panels represents the future of personalized medicine for brain tumors. Gathering current
data regarding biomarker candidates, we address the major challenges surrounding the
biomarker field of this devastating tumor type, exploring potential perspectives for the
development of more effective predictive biomarker panels.

The most common and lethal primary type of brain tumors is represented by high-grade gliomas; KEYWORDS 
they present one of the highest rates of occurrence within the tumors of the CNS. Grade IV glioma • biomarker panels • brain
– glioblastoma (GBM) – represents the most common and aggressive type of brain tumor in adults tumors • circulating
with a poor prognosis of a 1–2-year survival rate after diagnosis [1] . biomarkers • proteomics
The treatment of GBM is a significant therapeutic challenge; it is obvious that new approaches • targeted therapy
regarding classical and new therapeutic targets involving angiogenic signals, signaling pathways,
protein–protein interactions, stem cell targets and crosstalk between all of them are still an unmet
need in this disease.
Due to the high complexity and heterogeneity of this type of cancer, conventional biochemical
methods can fail to notice important components and/or proteomic profiles of this disease. One of
the main goals of oncology is represented by biomarkers development, since it presents the potential
to identify cancer risks, as well as to improve early detection and targeted therapy. We are confident
that a biomarker panel would provide superior identifiers and/or predictors of a patient’s clinical
outcome. High-throughput proteomic profiling and multiplex analysis, such as xMAP and protein
arrays technologies, are becoming important approaches in GBM research. These technologies can
supply simultaneous analyses of biomarkers in a panel for improved diagnosis, patient stratification,
prognosis and drug screening. Biomarker discovery for brain tumors is an ongoing pursuit and the
search for the best molecule or combination of molecules is still unfolding [2] . In the last 5 years,
proteomics topics were a major presence in the literature, having a bulk of more than 5000 papers

1
Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, no 99–101 Splaiul Independentei, 050096 Sector
5 Bucharest, Romania
2
National Institute for Chemical Pharmaceutical R&D, 112 Calea Vitan, 031299 Sector 3, Bucharest, Romania
3
Carol Davila University of Medicine & Pharmacy, Cell Biology & Histology Department, no 8 B-dul Eroilor Sanitari, 050474 Sector 5
Bucharest, Romania
4
Stefan S Nicolau Institute of Virology, Bucharest, Romania
*Author for correspondence: bioch@vbabes.ro

10.2217/FON.14.238 © 2015 Future Medicine Ltd Future Oncol. (2015) 11(3), 511–524 ISSN 1479-6694 511
Review  Tanase, Albulescu, Codrici et al.

focusing on proteomics in cancer; around half Loss of heterozygosity (LOH) on chromosome


of these papers report biomarker candidates in 1p/19q and chromosome 10 may help to predict
intracranial tumors. In spite of this abundance, patient outcomes [11] .
clinicians still do not count on a specific tumor The assessment of these specific alterations
marker for brain tumors. has been encumbered by the scarceness of biotic
Therefore, this review, along with our hands- material; therefore, their detection in the cir-
on experience, emphasizes the latest knowledge culation would decrease diagnostic time and
in circulatory biomarkers, the tumor-derived expedite the therapeutic decision. Apart from
blood-based biomarkers, circulating tumor cells, genetic circulating tumor DNA analysis, which
circulating nucleic acids and circulating proteins emulates tumor assessment for genetic and epi-
in patients with malignant gliomas. genetic aberrations, serum profiling for protein
biomarkers has also been proven useful as a diag-
Molecular classification of GBM nostic and/or prognostic tool for GBM. It is now
Histological and molecular analysis of CNS commonly accepted that a panel of biomarkers
tumors can identify approximately 120 sub- shows better predictive power over a single bio-
types. Peripheral blood scans for predictive marker, and recently, due to the development
biomarkers in CNS indicated that certain brain of proteomic techniques, panel attainment has
tumors are indeed associated with distinct pro- become a feasible task [12] . Hence, a number of
files of circulating factors, such as proteins (e.g., proteins has been identified as potential diag-
glial fibrillary acidic protein), DNA fragments nostic markers (A2M, factor VII, MDC, SCF
[3] or miRNAs (e.g., miRNA-21) [4] . [13] and staging biomarkers [haptoglobin, plas-
So far, the only subclassification of GBM minogen precursor, apolipoprotein A-1 and M,
that also inf luences the therapeutic deci- and transthyretin]) [14] . A systematic review of
sion is the detection of O-methylguanine- multiple independent proteomic analyses of
DNA methyltransferase (MGMT ) promoter glioma, carried out by Deighton et al., demon-
hypermethylation. strated alterations in 99 different proteins, ten of
Recent progress in the molecular research of which were repeatedly reported (PHB, Hsp20,
GBM has led to the identification of other mark- serum albumin, EGF receptor, EA-15, RhoGDI,
ers useful in either the diagnosis or prognosis of APOA1, GFAP, HSP70 and PDIA3) [15] .
this fatal disease. Mutation in isocitrate dehy-
drogenase genes, notably for isoform 1 (IDH1), Circulating tumor cells in malignant
seems to bear favorable prognostic value for GBM gliomas
patients [5] . Furthermore, The Cancer Genome Circulating tumor cells (CTCs), originating
Atlas Research Network showed evidence for a both from primary and metastatic lesions, can
CpG island methylator phenotype in GBMs, be detected in the peripheral blood of patients
associated with IDH1 mutations [6] . The newly with different solid tumors [16] . Unlike normal
emerged data involving IDH mutations and epi- blood cells, CTCs are present in very low counts,
genetic modifications prompted further inquir- up to a few hundred per milliliter, depending on
ies regarding the molecular mechanisms that the CTC definition and the methods used for
occur up- and down-stream from these events. their detection and isolation [17] .
RBP1 promoter hypermethylation is found in Several clinical studies performed in patients
nearly all IDH1 and IDH2 mutant gliomas, and with metastatic breast, prostate, colorectal and
is associated with improved patient survival [7] . non-small-cell lung cancer (NSCLC) showed
TET enzymes that catalyze oxidation of 5mC to a correlation between CTC count and clinical
5hmC and epigenetically modify gene transcrip- outcome, and suggested that CTC enumera-
tion are mutated in several types of cancer, affect- tion may represent a noninvasive adjuvant tool
ing their activity and likely altering genomic for prognosis, recurrence and therapy response
5hmC and 5mC patterns [8] . Modified expres- monitoring [18] .
sion of TET genes seems to have a significance However, it will be very useful in clinical
in risk stratification of GBM patients, along with practice to detect CTCs in patients with early-
APOBEC deaminase genes [9] . Promoter hyper- stage cancer in order to benefit from more
methylation of p16INK4, p14ARF, RB, PTEN adequate risk stratification and personalized
and p53 were also reported to play a role in the therapy. This approach is limited by the fact
epigenetic mechanisms of GBM pathology [10] . that most of the current methods are not able to

512 Future Oncol. (2015) 11(3) future science group


Circulating biomarker panels for targeted therapy in brain tumors  Review

accurately establish the CTC numbers in early- in the oncogenic transformation of various cells.
stage tumors, and further investigation is needed It has been recently noted that oncosomes can
for this purpose. contain several types of molecules involved in
Regarding malignant gliomas, although this pathogenesis pathways, such as tumor prolif-
type of tumor is thought to rarely disseminate eration, angiogenesis and invasion. Oncosomes
outside the brain, there are reports in the lit- can contain proteins, transcripts, DNA and
erature of lung, liver, lymph node and bone miRNAs that ‘hijack’ the recipient cell’s physi-
metastasis occurring in patients with GBM ology and cell microenvironment. Oncogenic
multiforme [19] . Moreover, some cases of GBM EGF receptor (EGFRvIII), tumor suppressors
transmitted through organ transplantation from (PTEN) and oncomirs (miR-520g) were found
affected donors are well documented [20] . These in the oncosomes identified in blood circulation
data provide indirect evidence that CTCs are and the cerebrospinal fluid (CSF) of patients
present in patients with malignant gliomas, diagnosed with brain tumors. These new ‘cir-
although thus far they have not been ­successfully culatory’ biomarkers could be useful biomark-
detected [21] . ers in patient stratification and/or therapeutic
Due to the fact that brain tumors, such as efficacy predictors [24] . As we are focusing on
GBM, tend to lack cancer cell surface biomark- circulatory biomarker candidates, the role of
ers such as EpCAM on which current methods exosomes/oncosomes in cancer, hence a circula-
of CTC detection are based, in a very recent tory particle capable of transferring tumor cell-
paper Macarthur KM et al. proposed a new derived genetic material and signaling proteins
strategy to detect CTCs. They used a telomer- from one cell to another and thus linking tumo-
ase-specific adenoviral agent to assess telomerase rigenesis, angiogenesis and metastasis, is even
activity, which is increased in nearly all tumor more intriguing. In 2013 it was reported that
cells, but not in normal cells. The assay proved internalization of exosomes was derived from
to successfully detect CTCs in patients with GBM cells where the protein CAV1 negatively
brain tumors, thus providing a promising tool regulates the uptake of exosomes. Moreover,
for monitoring treatment response [22] . exosomes can induce activation of ERK1/2 and
High-grade gliomas are characterized by HSP27, opening new opportunities for therapy
increased neovascularization and recruitment targets [25] .
of endothelial progenitor cells (EPCs); this rep- Thus, oncosomes have come into view in
resents one of the mechanisms involved in new brain tumor research domain as important vehi-
vessel formation. Related to this topic, Corsini cles of intercellular communication, providing a
E et al. studied the effect of surgical and post- new field of molecular biomarkers investigation.
surgical treatment at the level of EPCs in glioma
patients and their correlation with VEGF. They miRNAs as molecules linking tissue
found significantly decreased levels of EPCs in & circulating biomarkers
all treated patients compared with untreated The discovery of miRNA in tissues, as well as
ones. VEGF registered decreased levels only in body fluids together with their altered pro-
after surgery, but not after chemotherapy; no file in various pathological conditions, offers
correlation was found between VEGF and EPCs a new perspective on the use of extracellular
levels. Further studies are needed to assess the miRNAs as informative biomarkers of disease
usefulness of EPCs as markers of angiogenesis [26,27] . Similarly to other types of tumors, GBM
monitoring in high-grade gliomas [23] . multiforme was associated with numerous miR-
NAs. Such miRNAs display either oncogenic or
Oncosomes as future circulating tumor-suppressive properties, and are therefore
biomarkers carriers attractive therapeutic targets for future miRNA-
The complex interactions between CNS cell based therapies. Moreover, some studies have
types and distinctive cellular milieu, gener- suggested that miRNAs may be used as nonin-
ates a particular biology for brain tumors, vasive diagnostic and prognostic biomarkers due
which impacts on their sensitivity to treatment. to their release in the circulation [28] .
Oncosomes, or membrane-derived extracellu- In our previous paper, we also highlighted the
lar vesicles (EVs) secreted by cancer cells, can fact that miRNA expression profiles in GBM
transport proteins and nucleic acids from one are better predictors of clinical outcome than
cell to the other, thus being active participants mRNA profiles [29] .

future science group www.futuremedicine.com 513


Review  Tanase, Albulescu, Codrici et al.

In order to select candidate miRNA biomark- Another miRNA found to be downregulated


ers for malignant astrocytomas, Yang et al. per- in GBM is miR-34a; its expression has an anti-
formed genome-wide miRNA screening of serum tumor effect suppressing cell proliferation, G1/S
samples from astrocytoma patients and matched cell cycle progression, cell survival, cell migra-
controls. After subsequent validation of the tion and cell invasion in glioma cell lines. These
obtained results, seven miRNAs were found to be properties are due to inhibition of c-Met, Notch-1
significantly decreased in patients versus controls: and Notch-2 [39] .
miR-15b, miR-23a, miR-133a, miR-150, miR-
197, miR-497 and miR-548b-5p. Moreover, these Proteomic profiling for circulating
identified miRNAs exhibited a marked increase biomarkers discovery
after surgery. As a conclusion, this miRNA pro- During the last decade, a wide range of tech-
file may be a potential noninvasive biomarker nologies has been employed in molecular and
panel for malignant astrocytoma [30] . genetic profiling studies for biomarker discovery
Different studies have demonstrated abnor- in brain tumors. However, a limited number of
mal miRNA expression profiles in GBM tis- biomarkers has been identified and none has
sues; some miRNAs were found to be upreg- yet attained broad application for use in clinical
ulated (e.g., miR-17, miR-21, miR-93 and gliomas prognosis, therapeutic target selection
miR-221/222), while others were downregulated or molecular classification [40] .
(miR-7, miR-34a, miR-128 and miR-137) [31] . Proteomic profiling offers large-scale analysis
Lu et al. found an overexpression of miRNA- of protein expression, post-translational modi-
17 in glioma tissues that was associated with fication and protein–protein interactions. The
advanced pathological stage and poor overall ideal platforms for biomarker discovery include
survival [32] , while in a previous study conducted genomic and metabolomic analyses, and have
on glioma cell lines, Malzkorn et al. showed that the ultimate goal of unifying the information
miR-17 inhibition reduces cell viability and into protein networks [41] .
increases apoptotic activity [33] . While advanced proteomics, based on combi-
In vivo and in vitro studies on glioma cells nations of techniques such as 2D or 2D-DIGE
pointed out that miR-93-overexpressing cells and mass spectrometry, are the solution of choice
promoted endothelial cell spread, growth, for detailed studies that involve sequencing,
migration and tube formation supporting neo- high-throughput techniques, such as SELDI-
angiogenesis and tumor growth. This effect is at TOF and MALDI-TOF, are used for differen-
least partly explained by the fact that miRNA-93 tial protein signatures. The major advantage of
targets and silences integrin-β8, whose expres- SELDI-TOF mainly lies in the fast output of
sion is associated with decreased cell growth [34] . molecular signatures, yet it has to be usually fol-
Significant overexpression of miR-221/222 lowed by more in-depth studies (based on more
was detected in high-grade gliomas compared accurate MS/MS technology) in order to more
with low-grade gliomas, related to increased cell accurately identify the proteins with modified
invasion and poor prognosis by directly targeting expression [42] .
TIMP3 [35] . Identification of potential neoplasia diagnos-
Recent work reported decreased expression tic, prognostic, predictive or treatment-assessing
of miR-128, which correlates with aggressive biomarkers could allow differential proteomic
human glioma subtypes; miRNA-128 exerts profiling of brain tumor versus disease-free state
tumor suppression effects by targeting RTK for the detection and monitoring of pathology-
signaling [36] . Although miRNA-128 was found related changes. Specifically, proteomic analysis
to be downregulated in GBM tissue compared of different fluids is less invasive than biopsy
with normal brain tissue, Roth et al. detected and could provide potentially informative
increased expression levels of miRNA-128 in causes of origin and progression of brain tumor
blood cells of GBM patients [37] . ­pathologies [40] .
Kefas B et al. found a significantly reduced A number of studies have reported different
expression of miR-7 in human GBM tissue and individual serum biomarkers for GBM, such as
GBM cell lines, showing that miR-7 directly YKL-40 [43] , GFAP [44] , MMP-9 [45] , EGFR [46]
inhibits EGFR expression and suppresses Akt and CD14 [47] .
pathway activation, and thus decreases the Reyens et al. reported elevation of several
­viability and invasiveness of GBM cells [38] . inflammatory proteins (C-reactive protein, IL-6,

514 Future Oncol. (2015) 11(3) future science group


Circulating biomarker panels for targeted therapy in brain tumors  Review

TNF-α and sialic acid), coagulation factors arrays are more suitable to perform quantitative
(fibrinogen, endogen thrombin generation, pro- analysis in high-throughput conditions [2] .
thrombin fragments 1 and 2, and tissue factor) Reliable circulating biomarkers could sup-
and angiogenic factors (VEGF, soluble VEGF port the management of gliomas by facilitating
receptor 1 and thrombospondin-1) in the plasma neuroradiological differential diagnosis at initial
of GBM patients [48] . presentation, planning of surgical interventions
SELDI-TOF MS technology has been utilized or monitoring of the disease course.
in the protein profiling analysis of a variety of Tumor initiation and progression represent a
specimens derived from patients with brain complex process involving genomic mutations,
tumors for the discovery of biomarkers that microenvironmental factors and inflammatory
facilitate early diagnosis, establish tumor grade mediators. An active role in determining the
and predict therapeutic outcome. malignancy phenotype is owned by the tumor
Kumar et al., based on a combination 2-DE/ microenvironment. Both host cells and cancer
MS approach, observed ten differentially cells crosstalk via a large variety of soluble fac-
expressed proteins in the sera of patients with tors, whose effects determine the final outcome
GBM and validated haptoglobin a2 as serum of the tumorigenic process [53] .
marker associated with tumor growth and Inflammatory cells, cytokines/chemokines
migration in GBM [49] . and their receptors existing in tumors contrib-
In another study based on SELDI-TOF MS, ute to tumor growth, progression, metastasis
Petrik et al. discovered and validated AHSG as and immunosuppression. They regulate tumor
a predictive and prognostic biomarker for GBM. growth either directly by transformation, sur-
Thus, low serum levels of AHSG were correlated vival, proliferation and migration of cancer
with a short median survival rate (<3 months), cells, or indirectly by enhancing angiogenesis
with normal levels in serum being associated or recruiting leukocytes [54] . The “match that
with prolonged survival (>2 years) [50] . lights the fire” of cancer is genetic damage; in
In a preliminary study [51] using SELDI-TOF this context, some types of inflammation may
MS on serum samples, we identified a number of provide the “fuel that feeds the flames” [55,56] .
11 clusters with significant differences between Matrix metalloproteases (MMPs), important
GBM and controls (p < 0.05) out of a total of components of the tumor microenvironment and
152 clusters with m/z values 2–55 kDa; six clus- secreted by glioma cell lines, are responsible for
ters were overexpressed and five underexpressed sVE release. Soluble VE-cadherin in the blood
in GBM patients compared with control. might reflect VEGF activity at the tumor site.
Gautam et al. [52] , using an iTRAQ-based Analysis of glioma patient sera confirmed the
LC-MS/MS approach in GBM patients plasma, presence of sVE in the bloodstream, and sVE
have identified a total of 296 proteins, out of levels were significantly predictive of overall sur-
which 61 exhibited increased levels in the vival, irrespective of the histopathological grade
patient group. Altered levels of FTL, S100A9 of tumors [57] .
and CNDP1 were validated by ELISA. These Plasma MMP-2 and VEGF were identified
proteins may form useful starting points for the in the study of Xu et al. as potential biomarkers
development of plasma-based biomarker panels that accurately distinguished high-grade glioma
in clinical investigations for GBM [52] . patients from controls [13] . MMP-2 and VEGF
have also been investigated in urine samples
Soluble biomarkers – main actors in panel as diagnostic biomarkers for brain tumors [58] .
set up MMP-2 promotes cell invasion, angiogenesis,
While mass spectrometry-based approaches are activation of growth factors, and is involved in
undoubtedly some of the most effective tools the development of human glioma microves-
in the de novo discovery of biomarkers, other sels, facilitating glioma cell invasion [59] . VEGF
approaches are available for sorting out biomark- can promote tumorigenesis and angiogenesis of
ers from known molecules, such as cytokines, human GBM stem cells, and VEGF expression is
chemokines or growth factors via protein arrays significantly increased in high-grade ­astrocytomas
or, more effective, xMAP multiplex technology. compared with low-grade astrocytomas [13] .
While protein arrays are generally more pow- Serum S100B and NSE were considered
erful in terms of the total number of proteins markers of CNS damage, yet further studies
that can be simultaneously detected, xMAP were required to establish whether S100B is an

future science group www.futuremedicine.com 515


Review  Tanase, Albulescu, Codrici et al.

independent prognostic biomarker in glioma An important issue regarding antiangio-


patients [60] . Subsequent studies have showen genic treatment discontinuation and patient
that biomarkers such as S100B, NPY and SCGN morbidity remains the toxicity derived from
were markedly elevated in plasma of patients antiangiogenic therapy, thus serum/plasma bio-
with malignant glioma 1 year before clinical marker panels that correlate with drug toxicity
manifestation [61] . and changes in cytokine and angiogenic fac-
Changes in serum levels of PlGF were associ- tors would be of great value. Changes in IL-13
ated with overall survival in patients with recur- from baseline to 24 h predicted on-target tox-
rent GBM [62] . Plasma concentrations for PlGF icities. Changes in IL-6, IL-10 and IL-13 were
were correlated to neuropathological or neuro- frequently correlated with toxicity. Profiling of
radiological features. In addition, IL-8, BDNF IL-13 as a surrogate for endothelial dysfunc-
and GDNF were detectable in different concen- tion could individualize patients at risk during
tration levels in serum/plasma. Measurement of antiangiogenic therapy [70] .
circulating GFAP and PlGF are potentially use- In our previous study [2] , a panel of cytokines
ful as clinical biomarkers that may support dif- with modified expression was detected in GBM
ferential diagnosis of GBM versus intracerebral patients’ sera using multiplex assay. Our results
metastasis [63] . indicate significant dysregulation in serum lev-
Serum IL-10 levels have been described as els of cytokines and angiogenic factors, three-
significantly increased in high-grade glioma fold upregulation for IL-6, IL-1β, TNF-α and
patients compared with nontumor control IL-10, and twofold upregulation of VEGF,
patients [55,64] . Increased levels of IL-10 in FGF-2, IL-8, IL-2 and GM-CSF were noticed
GBM patients are consistent with the findings (Figure 1) . Cytokines expression was strongly cor-
that Th2 cytokines are elevated in patients with related with tumor grade, proliferation markers
advanced tumors [13] . IL-10 was identified as a and clinical aggressiveness in GBMs [2] .
serum biomarker that accurately differentiates A circulating biomarker-based signature is an
GBM patients from controls [13] . important approach for the clinical assessment
A2M has been upregulated in migrating of malignant glioma, with the abovementioned
human glioma cells compared with nonmigrat- biomarkers facilitating accurate diagnosis and
ing glioma cells [65,66] . Neuronal and glioma- therapy monitoring, and assessing survival rate
derived SCF can induce angiogenesis within the prognosis [40] .
brain [67] . Expression of tissue factor, the cell Assessing circulating biomarkers through the
surface receptor for factor VII, correlates with rapid and efficient method represented by array
histologic grade of human glioma malignancy technology can be a reliable tool for the diagno-
and vascularity [68] . In addition to the accurate sis of brain tumors and for the discovery of new
classification of malignant glioma patients, cir- potential therapeutic targets or therapy monitor-
culating factor VII was also found to have prog- ing. Biomarker profiling, obtained via proteomic
nostic significance. MDC was underexpressed high-throughput technologies, could offer major
in plasma/serum samples from high-grade support to identify unique differences within an
glioma patients. MDC and its receptor CCR4 individual tumor or between tumors of the same
both show decreased expression in human grade histological grade [71] .
III astrocytoma and grade IV glioma cells [69] .
Knowing that these biomarkers have differential Signaling molecules profile as potential
expression levels in high-grade glioma tissues, biomarkers & therapy targets
they were also identified in glioma patients’ As stated in the previous sections, the aggres-
blood using multiplex assay [13] . Thus, A2M, fac- siveness and therapeutic resistance of GBM also
tor VII, MDC and SCF were identified as bio- represents the base for the search for signaling
markers in patients with malignant glioma, both dysregulation.
in plasma and serum specimens, with signifi- The intracellular pathways and the molecules
cant differences in the protein expression levels that append to these pathways could assume
between patients and controls. These molecules both the biomarker and future therapy target
represent protein biomarkers that may identify role [72] . GBM shows dysregulation in various
patients with malignant glioma in independent signaling pathways, including the G1/S cell cycle
cohorts with similar accuracy in either plasma checkpoint and the MAPK and PI3K effector
or serum specimens [13] . arms of RTK signaling [73] .

516 Future Oncol. (2015) 11(3) future science group


Circulating biomarker panels for targeted therapy in brain tumors  Review

Modulation of serum cytokine levels


in glioblastoma patients

IL-6
IL-1β
TNF-α
VEGF
FGF-2
IL-8
IL-2
GM-CSF
IFN-γ
IL-4
IL-12
0 1 2 3 4 5 6
Fold modification versus control

Figure 1. Modulation of serum cytokine levels in glioblastoma patients. The data represent group
averages of fold modification versus controls.

Dysregulations of PI3K/Akt/mTOR signaling PI3K inhibitor we have recorded upon cellular


were reported in GBM and induce RTK overac- responses is coherent with other experimen-
tivity (EGFR, PDGFR, mesenchymal–epithelial tal results pointing out the major relevance of
transition factor), mutated PI3K subunits and/or PI3K signaling in GBM and furthermore that
loss of tumor suppressor activity, namely PTEN PI3K inhibitors can add to, or even increase, the
[74] . Indeed, 40% of GBM have a loss of PTEN, limited success of EGFR inhibitors in clinical
triggering excessive PI3K signaling, while other trials [78] . P70S6K, a serine/threonine kinase,
GBMs exhibit post-translational modifications can be activated by both the PI3K and ERK
inducing inactivation of PTEN. A therapeuti- pathways. This multiple involvement supports
cally wise approach and more specific to person- the idea of the simultaneous usage of PI3K and
alized medicine can benefit from the identifica- MAPK inhibitors [79] . In accordance with our
tion of signaling dysregulation and the definition results, a similar interference between PI3K and
of novel targets [75] . The PI3K signaling pathway MEK/ERK signaling pathways was reported by
has been investigated as an important target for Sunayama et al. [80] .
the treatment of GBM [76] , seeking PI3K inhibi- Pan PI3K inhibitor 2-(4-morpholinyl)-8-phe-
tors as therapeutic agents in GBM [77] . According nyl-4H-1 benzopyran-4-one (LY294002) can
to one of our recently published papers, by induce important antitumoral overall effects in
inhibiting the PI3K pathway in patient-derived experimental models, but the LY294002 com-
GBM cells, the expression of signaling proteins pound has poor pharmacologic variables of insol-
belonging to various pathways can be altered, ubility and a short half-life. Starting from this
and hence their tumor cell proliferation capacity structure, a novel RGDS-conjugated LY294002,
can be reduced. Our recent study has revealed named SF1126, was designed. The goal was to
that treating GBM cell cultures isolated from exhibit increased solubility and bind to specific
patient tumors with PI3K inhibitors induced intratumor integrins, thus an cause enhanced
a significant decrease in the expression level of delivery of the active compound. Using the
several key signaling molecules involved in cell U87MG GBM cell line SF1126 had an enhanced
survival (p38), proliferation (ERK1/2, IκBα, efficacy due to the RGDS integrin (alpha v beta
p38 MAPK CREB), differentiation (ERK1/2, 3/alpha 5 beta 1) binding component. SF1126
CREB), migration (ERK1/2, CREB) and apop- has both antitumor and antiangiogenic activity,
tosis (ERK1/2, P70S6K, IκBα, JNK, CREB) [2] . recommending it as a viable pan PI3K inhibitor
We found that PI3K inhibitors reduced tumor for Phase I clinical trials [81] .
cell proliferation, as in similar reports for other In GBM tumor samples the axonal guidance
types of tumors [77] . The maximal efficacy of protein, netrin-1, is overexpressed. Last year it

future science group www.futuremedicine.com 517


Review  Tanase, Albulescu, Codrici et al.

was reported that, using tandem affinity puri- targets through CAV1 expression and ERK1/2
fication and mass spectrometry technologies, ­signaling [87] .
netrin-1 forms a complex with Notch2 and Intracellular signaling molecules that basi-
Jagged1. This co-localization was present on cally depict the molecular heterogeneity of the
the cell surface, while in intracellular vesicles tumor are certainly important biomarkers in
netrin-1 was present only with Jagged1, but not predicting responsiveness and disease outcome
with Notch2. Netrin-1 induced Notch signaling upon therapy. This assertion was sustained by
and consequent GBM invasion. Interestingly, less favorable clinical trial results when mono-
besides a candidate biomarker netri-1 can also therapy was used, therapy overlooking the com-
be a therapeutic target since the central domain plexity of intracellular network of individual
of netrin-1 counteracts the effects of netrin-1, tumors. Thus, intracellular key signaling mol-
meaning that it inhibits GBM cell invasion and ecules and the actual panel of these molecules are
Notch activation. This astonishing finding was to become important target therapies and valu-
that Notch signaling complex remained at the able biomarkers, especially for therapy ­efficacy
cell surface [82] . This year, a series of early-phase monitoring.
clinical trials were reported where Notch sign- The simultaneous detection of several mol-
aling was the therapy target using agents that ecules involved in various signaling pathways has
either obstructed Notch receptor cleavages such not been previously reported in GBM. Further
as γ-secretase inhibitors (GSIs) or interfered with studies are needed in order to assess whether
the Notch ligand–receptor interaction, which this regulation is transcriptional or post-tran-
occurs in several solid tumors, including intrac- scriptional. Keeping in mind that personalized
ranial ones [83,84] . This report again underlies medicine should be tailored to the patient’s
the urgent need for efficacy biomarkers to sup- tumor particularities, screening cell behav-
port the development of this class of drugs [85] . ior and expression levels of signaling proteins
Niclosamide induced the cytostatic, cytotoxic within tumor cells can highlight the important
and antimigratory effects of GBM in in vitro differences between primary tumors of similar
and in vivo models, and moreover reduced the histological type, and/or allow comparisons of
capacity of multipotent/self-renewing cells in primary and relapse tumor samples.
vitro. By analyzing the pathways triggered by
this drug, simultaneous inhibition of WNT/ Cancer stem cells as potential biomarkers
CTNNB1-, NOTCH-, mTOR- and NF-κB for GBM aggressiveness
cascades are revealed. The authors report that in Cancer stem cells (CSCs), identified in a myriad
GBM biological samples a heterozygous deletion of human solid tumors including brain tumors
of the NFκBIA locus was found, thus besides [88] , have generated a new field of research where
a possible genomic biomarker, this could serve a clear distinction should be made between can-
as an explanation for predicting the synergistic cer-initiating cells and cells that allow propaga-
activity of niclosamide with temozolomide, the tion of the tumor [89] . Nowadays, genomics and
current standard in GBM [75] . epigenetic techniques have proven that cancer
RTK-targeted therapy was investigated using takes control of specific genetic and epigenetic
imatinib, sunitinib and cediranib in GBM mod- programs from embryonic development circuits.
els. In a panel of ten GBM cell lines, cediranib Two recognized models regarding the genera-
proved to have the most potent antitumoral tion and function of GBM CSCs (GCSCs),
effect, while cediranib and sunitinib sensitizes the stochastic and the hierarchical models, are
the cells to the classic temozolomide. Cediranib ­currently recognized (Figure 2) .
inhibited MAPK and AKT pathways, but the According to the stochastic model, differenti-
authors did not find any correlation between ated or committed cells acquire genetic muta-
KIT, PDGFRA and VEGFR2 expression, and tions towards immortal proliferative capacity,
therapy responses to any of the RTK inhibitors. leading to cancer stem cells (GCSC in our
RTK therapy can rely upon future biomarkers case), in contrast with the hierarchical model
for therapy response in GBM [86] . where a stem cell or a glial precursor undergoes
As already stated in the previous section, a neoplastic transformation, leading to glioma
circulating exosomes can induce the activa- initiation and development [90] . Properties such
tion of ERK1/2 and HSP27 via CAV1; this as extensive self-renewal ability and multipotency
recent report opens new possibilities for therapy are common for neural stem cells (NSCs) and

518 Future Oncol. (2015) 11(3) future science group


Circulating biomarker panels for targeted therapy in brain tumors  Review

T
NSC CSC
Muta
tion

CSC
on
Mutati
NSC PC Mutati
on

CPC

DC

Several mutation events

Figure 2. Acknowledged theories regarding cancer stem cell generation – stochastic and
hierarchical theories. (A) Normal neural stem cells (NSCs) give rise to progenitors cells (PCs) with
limited proliferative capacity that further leads to differentiated normal cells (DCs). CSCs can form as
a result of abnormal differentiation of a normal NSC or neural progenitor cell (PC). CSC can develop
cancer progenitor cells (CPCs) that further are able to develop the actual tumor (T). (B) CSCs can
result from terminally differentiated cells, which acquire several genetic mutations.

GCSCs, and have been included in the defini- As mentioned in the previous section, the
tion of cancer stem cells. It has recently been Notch pathway is highly activated in this type of
shown that chemokine CXCL12 and its recep- solid tumor. Moreover, it has recently been shown
tor CXCR4 can control proliferation, invasion in an animal model that mice constitutively
and angiogenesis in GBM cell lines and primary expressing the activated intracellular domain
cultures. In GCSCs an increased CXCR4 expres- of Notch2 display neurogenic niche hyperpla-
sion was found, as well as release of CXCL12 sia and reduced neuronal lineage entry. When
in vitro. As also found by us, there is a clear het- authors isolated neurospheres from this model,
erogeneity of GBMs that induce different levels an increased proliferation, survival and resistance
of both expression and secretion in individual to apoptosis was obtained. In human GBM cell
cultures, again an important argument for per- lines, the Notch2 pathway induces an increased
sonalized therapy. CXCL12 activation-induced proliferation and resistance to apoptosis. When
Akt-mediated reduced apoptosis and self-renewal assessing gene expression in GBM patient tumor
activities, but less proliferation, while CXCR4 samples, a positive correlation of Notch2 tran-
antagonist AMD3100 reduced self-renewal and scripts with the transcripts that control antia-
survival. In in vitro differentiated cells derived poptotic processes, stemness and astrocyte fate
from the same GBMs, the abovementioned inhi- was found. In the meantime, Notch2 transcripts
bition through AMD3100 was not obtained, this were negatively correlated with gene transcripts
finding being a strong argument for the coupled controlling the proapoptotic process and oligo-
CXCL12/CXCR4 interactions that are specific dendrocyte fate. The Notch2 pathway in NSCs
for CSC in GBM mediating survival and self- can drive to GCSCs and can induce astrocytic
renewal, therefore representing a promising lineage entry and brain tumor ­development [92] .
future for therapeutic targets [91] . We point out GCSC autophagy was reported to be induced
that this finding can be also extrapolated to cir- by suberoylanilide hydroxamic acid (SAHA),
culatory biomarkers, as CXCL12 is secreted by a histone deacetylase inhibitor, as an effect of
GCSCs, enhancing the possibility to find this late-phase apoptosis. In vivo xenografts have
biomarker in patients’ circulation. shown that SAHA reduced tumor growth and

future science group www.futuremedicine.com 519


Review  Tanase, Albulescu, Codrici et al.

determined autophagy. Actually, the authors TrkA, TrkB and TrkC) and their ligands (NGF,
report that SAHA’s action is induced by down- BDNF and NT3) were reported. Moreover,
regulation of AKT/mTOR signaling. Taken BTICs secrete NGF and, thus, exogenouous
together, this recent study shows that in GCSC NGF induced BTIC proliferation. The authors
therapeutic approaches, SAHA can be a capable note that the intracellular domain of p75NTR is
agent for autophagy induction [93] . to be found in GBM biological specimens, sug-
This year, in brain tumor initiating cells gesting that the receptor is activated and cleaved
(BTICs), neurotrophin receptors (p75NTR, in patient tumors. These results open a new

EXECUTIVE SUMMARY
Molecular classification of glioblastoma
●● Histologic and molecular analysis of CNS tumors can identify approximately 120 subtypes.
●● Recent progress in molecular research of GBM has led to the identification of novel biomarkers useful in the diagnosis
and prognosis of this fatal disease.
Circulating tumor cells in malignant gliomas
●● Circulating tumor cells originating from both primary and metastatic lesions can be detected in the peripheral blood
of patients with different solid tumors.
●● Several clinical studies performed in patients with different types of tumors, including brain tumors, showed a
correlation between CTCs and clinical outcome, and suggested that CTC detection may represent a noninvasive
adjuvant tool for evaluating prognostic value, recurrence capacity and therapy response monitoring.
Oncosomes as a future circulating biomarkers carrier
●● Oncosomes, or membrane-derived extracellular vesicles (EVs), secreted by cancer cells can transport proteins and
nucleic acids from one cell to the other, thus being active participants in the oncogenic transformation of various cells.
●● Oncosomes come into view in brain tumor research domains as an important vehicle of intercellular communication,
providing a new field of molecular biomarkers investigation.
miRNAs as molecules linking tissue & circulating biomarkers
●● Tumor-specific miRNAs are promising tools for the early detection of cancer and the development of personalized
therapies.
Proteomic profiling for circulating biomarkers discovery
●● A wide range of technologies have been employed in molecular profiling studies for biomarker discovery in brain
tumors. Proteomic profiling offers large-scale analysis of protein expression, post-translational modification and
protein–protein interactions.
●● SELDI-TOF-MS technology has been utilized in protein profiling analysis for the discovery of biomarkers facilitating
diagnosis, predicting therapeutic response and establishing tumor grade.
Signaling molecule profile with biomarker potency & future therapy targets
●● GBM shows dysregulation in various signaling pathways including the G1/S cell cycle checkpoint and the MAPK and
PI3K effectors of RTK signaling.
●● Deregulations of PI3K/Akt/mTOR signaling were reported in GBM and induce RTK overactivity (EGFR, PDGFR), mutated
PI3K subunits and/or loss of tumor suppressor activity, namely PTEN.
Cancer stem cells as potent biomarkers for GBM aggressiveness
●● Cancer stem cells, identified in a myriad of human solid tumors including brain tumors, have generated a new field of
research where a clear distinction should be made between cancer-initiating cells and cells that allow propagation of
the tumor.
●● Several molecules have been identified as candidate biomarkers for glioblastoma, but a more suitable approach for
efficacious therapy would rely on the use of personalized proteomic profiles.

520 Future Oncol. (2015) 11(3) future science group


Circulating biomarker panels for targeted therapy in brain tumors  Review

door in the BTIC research domain for a novel Future therapies could emerge from the coopera-
­potential clinical target [94] . tion between cancer stem cells and their niches,
which will improve the maintenance of their
Conclusion & future perspective characteristic features and behavior, with the
Although there are many proteins described concomitant activation of differentiation signal-
as ‘potential biomarkers’ and although predic- ing pathways, such as RTKs–Akt, Notch, BMPs,
tive biomarkers have not been yet validated for Hedgehog, Wnt-β-catenin, and so on.
patients with GBM, a useful serum marker panel New protocols based on a combination of
of brain tumors is urgently needed. Discovering chemotherapy and immunotherapy are probably
a single biomarker that would be both sensi- a new approach for achieving therapeutic syn-
tive and specific for cancer might be more dif- ergy and more suitability for brain tumors. The
ficult than discovering a panel of biomarkers. cancer stem cell paradigm might become the
Although some molecules have been proposed cornerstone for novel cancer research approaches
as potential GBM biomarkers, it is the current in the following years.
opinion that a more adequate approach would In this review, we have addressed the major
be to compare the proteomic profile of an indi- challenges surrounding biomarker development
vidual with the values recorded in healthy indi- in this particular type of devastating tumors; by
viduals (panel of biomarkers). gathering the current data regarding biomarker
We believe that in the very near future a panel candidates, we can explore potential routes for
comprising cytokines, chemokines and angio- the development of a more effective predictive
genic circulating biomarkers can offer an insight biomarkers panel.
regarding the diagnostic and invasive potential
of GBM. This front panel will be followed by Financial & competing interests disclosure
the identification of CTC along with miRNA This paper is partly supported by the Sectorial Operational
panels that depict disease progression, relapse Programme Human Resources Development (SOPHRD),
and therapy monitoring. In the long term, it is and financed by the European Social Fund and the Romanian
a probability that intracellular signaling path- Government under the contract number POSDRU 141531.
ways elucidation accompanied by circulating This work was partially supported by grants PN 09.33-04.15,
oncosomes will be developments in the bio- PN 09.33-03.10 and PN 09.33-01.01. The authors would
markers field, providing this pathology with like to thank Irina Radu, certified translator in Medicine
new therapy targets. – Pharmacy, certificate credentials: series E no. 0048, for
GBM patients will benefit more from person- professional linguistic assistance. The authors have no other
alized therapy by tailoring their treatment and relevant affiliations or financial involvement with any
aiming specific protein–protein interactions and organization or entity with a financial interest in or financial
signaling networks according to tumor pheno- conflict with the subject matter or materials ­discussed in the
type or patient clustering based on biomarker manuscript apart from those disclosed.
panels. New, promising fields for GBM personal- No writing assistance was utilized in the production of
ized medicine are microRNA and CSC signaling. this manuscript.

References 4 Ilhan-Mutlu A, Wagner L, Wohrer A et al. 7 Chou AP, Chowdhury R, Li S et al.


Papers of special note have been highlighted as: Plasma MicroRNA-21 concentration may Identification of retinol binding protein 1
• of interest; •• of considerable interest be a useful biomarker in glioblastoma promoter hypermethylation in isocitrate
patients. Cancer Invest. 30(8), 615–621 dehydrogenase 1 and 2 mutant gliomas.
1 Louis DN, Ohgaki H, Wiestler OD et al. The
(2012). J. Natl Cancer Inst. 104(19), 1458–1469
2007 WHO classification of tumours of the
5 Sanson M, Marie Y, Paris S et al. Isocitrate (2012).
central nervous system. Acta Neuropathol.
114(2), 97–109 (2007). dehydrogenase 1 codon 132 mutation is an 8 Kinney SR, Pradhan S. Ten eleven
important prognostic biomarker in gliomas. translocation enzymes and
2 Cruceru ML, Enciu AM, Popa AC et al. Signal
J. Clin. Oncol. 27(25), 4150–4154 (2009). 5-hydroxymethylation in mammalian
transduction molecule patterns indicating
•• Of outstanding importance for advancement development and cancer. Adv. Exp. Med. Biol.
potential glioblastoma therapy approaches.
754, 57–79 (2013).
Onco. Targets Ther. 6, 1737–1749 (2013). in prognostic of gliomas
9 Orr BA, Haffner MC, Nelson WG,
3 Lavon I, Refael M, Zelikovitch B, Shalom E, 6 Muller T, Gessi M, Waha A et al. Nuclear
Yegnasubramanian S, Eberhart CG.
Siegal T. Serum DNA can define tumor- exclusion of TET1 is associated with loss of
Decreased 5-hydroxymethylcytosine is
specific genetic and epigenetic markers in 5-hydroxymethylcytosine in IDH1 wild-type
associated with neural progenitor phenotype
gliomas of various grades. Neuro. Oncol. gliomas. Am. J. Pathol. 181(2), 675–683
in normal brain and shorter survival in
12(2), 173–180 (2010). (2012).

future science group www.futuremedicine.com 521


Review  Tanase, Albulescu, Codrici et al.

malignant glioma. PLoS ONE 7(7), e41036 gliomas. J. NeuroOncol. 113(3), 345–352 predicts poor prognosis in human glioma.
(2012). (2013). J. BioMed. Biotechnol. 2012, 970761 (2012).
10 Alelu-Paz R, Ashour N, Gonzalez-Corpas A, 22 Macarthur KM, Kao GD, Chandrasekaran S 33 Malzkorn B, Wolter M, Liesenberg F et al.
Ropero S. DNA methylation, histone et al. Detection of brain tumor cells in the Identification and functional characterization
modifications, and signal transduction peripheral blood by a telomerase promoter- of microRNAs involved in the malignant
pathways: a close relationship in malignant based assay. Cancer Res. 74(8), 2152–2159 progression of gliomas. Brain Pathol. 20(3),
gliomas pathophysiology. J. Signal. Transduct. (2014). 539–550 (2010).
2012, 956–958 (2012). 23 Corsini E, Ciusani E, Gaviani P et al. 34 Fang L, Deng Z, Shatseva T et al. MicroRNA
11 Mizoguchi M, Kuga D, Guan Y et al. Loss Decrease in circulating endothelial progenitor miR-93 promotes tumor growth and
of heterozygosity analysis in malignant cells in treated glioma patients. J. NeuroOncol. angiogenesis by targeting integrin-beta8.
gliomas. Brain Tumor Pathol. 28(3), 191–196 108(1), 123–129 (2012). Oncogene 30(7), 806–821 (2011).
(2011). 24 D’asti E, Garnier D, Lee TH, Montermini L, 35 Zhang C, Zhang J, Hao J et al. High level of
12 Tumilson CA, Lea RW, Alder JE, Shaw L. Meehan B, Rak J. Oncogenic extracellular miR-221/222 confers increased cell invasion
Circulating microRNA biomarkers for glioma vesicles in brain tumor progression. Front. and poor prognosis in glioma. J. Transl. Med.
and predicting response to therapy. Mol. Physiol. 3, 294 (2012). 10, 119 (2012).
Neurobiol. doi:10.1007/s12035-014-8679-8 • Of interest for understanding extracellular 36 Papagiannakopoulos T, Friedmann-Morvinski
(2014) (Epub ahead of print). D, Neveu P et al. Pro-neural miR-128 is a
vesicles as “a novel and functionally
13 Xu BJ, An QA, Srinivasa Gowda S et al. important vehicle of intercellular glioma tumor suppressor that targets mitogenic
Identification of blood protein biomarkers communication that can mediate multiple kinases. Oncogene 31(15), 1884–1895 (2012).
that aid in the clinical assessment of patients biological effects.” 37 Roth P, Wischhusen J, Happold C et al. A
with malignant glioma. Int J. Oncol. 40(6), specific miRNA signature in the peripheral
25 Svensson KJ, Christianson HC, Wittrup A
1995–2003 (2012). blood of glioblastoma patients. J. Neurochem.
et al. Exosome uptake depends on ERK1/2-
14 Gollapalli K, Ray S, Srivastava R et al. heat shock protein 27 signaling and lipid 118(3), 449–457 (2011).
Investigation of serum proteome alterations in Raft-mediated endocytosis negatively • Of interest for advancement in diagnostic of
human glioblastoma multiforme. Proteomics regulated by caveolin-1. J. Biol. Chem. glioblastoma patients
12(14), 2378–2390 (2012). 288(24), 17713–17724 (2013).
38 Kefas B, Godlewski J, Comeau L et al.
15 Deighton RF, Mcgregor R, Kemp J, 26 Moldovan L, Batte KE, Trgovcich J, Wisler J, microRNA-7 inhibits the epidermal growth
Mcculloch J, Whittle IR. Glioma Marsh CB, Piper M. Methodological factor receptor and the Akt pathway and is
pathophysiology: insights emerging from challenges in utilizing miRNAs as circulating down-regulated in glioblastoma. Cancer Res.
proteomics. Brain Pathol. 20(4), 691–703 biomarkers. J. Cell Mol. Med. 18(3), 371–390 68(10), 3566–3572 (2008).
(2010). (2014).
39 Guessous F, Zhang Y, Kofman A et al.
16 Yu M, Stott S, Toner M, Maheswaran S, 27 Wang J, Zhang KY, Liu SM, Sen S. microRNA-34a is tumor suppressive in brain
Haber DA. Circulating tumor cells: Tumor-associated circulating microRNAs as tumors and glioma stem cells. Cell Cycle 9(6),
approaches to isolation and characterization. biomarkers of cancer. Molecules 19(2), 1031–1036 (2010).
J. Cell Biol. 192(3), 373–382 (2011). 1912–1938 (2014).
40 Kalinina J, Peng J, Ritchie JC, Van Meir EG.
17 Van De Stolpe A, Pantel K, Sleijfer S, 28 Odjele A, Charest D, Morin P Jr. miRNAs as Proteomics of gliomas: initial biomarker
Terstappen LW, Den Toonder JM. important drivers of glioblastomas: a discovery and evolution of technology. Neuro.
Circulating tumor cell isolation and no-brainer? Cancer Biomark. 11(6), 245–252 Oncol. 13(9), 926–942 (2011).
diagnostics: toward routine clinical use. (2012).
Cancer Res. 71(18), 5955–5960 (2011). 41 Choudhary C, Mann M. Decoding signalling
29 Tanase CP, Enciu AM, Mihai S, Neagu AI, networks by mass spectrometry-based
18 Hou JM, Greystoke A, Lancashire L et al. Calenic B, Cruceru ML. Anti-cancer proteomics. Nat. Rev. Mol. Cell Biol. 11(6),
Evaluation of circulating tumor cells and therapies in high grade gliomas. Curr. 427–439 (2010).
serological cell death biomarkers in small cell Proteomics 10(3), 246–260 (2013).
lung cancer patients undergoing 42 Popescu ID, Albulescu R, Raducan E,
30 Yang C, Wang C, Chen X et al. Identification Dinischiotu A, Tanase CP. Applications
chemotherapy. Am. J. Pathol. 175(2),
of seven serum microRNAs from a genome- of SELDI-TOF technology in cancer
808–816 (2009).
wide serum microRNA expression profile as biomarkers discovery. 15(5), 5654–5667
19 Taskapilioglu MO, Aktas U, Eser P, Tolunay potential noninvasive biomarkers for (2010).
S, Bekar A. Multiple extracranial metastases malignant astrocytomas. Int. J. Cancer
from secondary glioblastoma: a case report 43 Iwamoto FM, Hottinger AF, Karimi S et al.
132(1), 116–127 (2013).
and review of the literature. Turk. Neurosurg. Serum YKL-40 is a marker of prognosis
31 Moller HG, Rasmussen AP, Andersen HH, and disease status in high-grade
23(6), 824–827 (2013).
Johnsen KB, Henriksen M, Duroux M. A gliomas. Neuro. Oncol. 13(11), 1244–1251
20 Fatt MA, Horton KM, Fishman EK. systematic review of microRNA in (2011).
Transmission of metastatic glioblastoma glioblastoma multiforme: micro-modulators
multiforme from donor to lung transplant 44 Gallego Perez-Larraya J, Paris S, Idbaih A
in the mesenchymal mode of migration and
recipient. J. Comput. Assist. Tomogr. 32(3), et al. Diagnostic and prognostic value of
invasion. Mol. NeuroBiol. 47(1), 131–144
407–409 (2008). preoperative combined GFAP, IGFBP-2, and
(2013).
YKL-40 plasma levels in patients with
21 Holdhoff M, Yovino SG, Boadu O, Grossman 32 Lu S, Wang S, Geng S, Ma S, Liang Z, Jiao B. glioblastoma. Cancer doi:10.1002/cncr.28949
SA. Blood-based biomarkers for malignant Increased expression of microRNA-17 (2014) (Epub ahead of print).

522 Future Oncol. (2015) 11(3) future science group


Circulating biomarker panels for targeted therapy in brain tumors  Review

45 Iwamoto FM, Hottinger AF, Karimi S et al. towards a candidate biomarker. PLoS ONE 70 Shonka N, Piao Y, Gilbert M et al. Cytokines
Longitudinal prospective study of matrix 8(12), e80056 (2013). associated with toxicity in the treatment of
metalloproteinase-9 as a serum marker in 58 Smith ER, Zurakowski D, Saad A, Scott RM, recurrent glioblastoma with aflibercept. Target
gliomas. J. Neurooncol. 105(3), 607–612 Moses MA. Urinary biomarkers predict brain Oncol. 8(2), 117–125 (2013).
(2011). tumor presence and response to therapy. Clin. 71 Rosser CJ, Goodison S. Today’s discoveries to
46 Quaranta M, Divella R, Daniele A et al. Cancer Res. 14(8), 2378–2386 (2008). tomorrow’s care: cancer biomarkers revisited.
Epidermal growth factor receptor serum levels 59 Girolamo F, Virgintino D, Errede M et al. Foreword. Biomark. Med. 4(4), 491–493
and prognostic value in malignant gliomas. Involvement of metalloprotease-2 in the (2010).
Tumori 93(3), 275–280 (2007). development of human brain microvessels. 72 Van Meir EG, Hadjipanayis CG, Norden AD,
47 Zhou M, Wiemels JL, Bracci PM et al. Histochem. Cell Biol. 122(3), 261–270 Shu HK, Wen PY, Olson JJ. Exciting new
Circulating levels of the innate and humoral (2004). advances in neuro-oncology: the avenue to a
immune regulators CD14 and CD23 are 60 Vos MJ, Postma TJ, Martens F et al. Serum cure for malignant glioma. CA Cancer J. Clin.
associated with adult glioma. Cancer Res. levels of S-100B protein and neuron-specific 60(3), 166–193 (2010).
70(19), 7534–7542 (2010). enolase in glioma patients: a pilot study. 73 Wieland A, Trageser D, Gogolok S et al.
48 Reynes G, Vila V, Martin M et al. Circulating Anticancer Res. 24(4), 2511–2514 (2004). Anticancer effects of niclosamide in human
markers of angiogenesis, inflammation, and 61 Gartner W, Ilhan A, Neziri D et al. Elevated glioblastoma. Clin. Cancer Res. 19(15),
coagulation in patients with glioblastoma. blood markers 1 year before manifestation of 4124–4136 (2013).
J. NeuroOncol. 102(1), 35–41 (2011). malignant glioma. Neuro. Oncol. 12(9), 74 Fenton TR, Nathanson D, Ponte De
49 Kumar DM, Thota B, Shinde SV et al. 1004–1008 (2010). Albuquerque C et al. Resistance to EGF
Proteomic identification of haptoglobin 62 Batchelor TT, Duda DG, Di Tomaso E et al. receptor inhibitors in glioblastoma mediated
alpha2 as a glioblastoma serum biomarker: Phase II study of cediranib, an oral by phosphorylation of the PTEN tumor
implications in cancer cell migration and pan-vascular endothelial growth factor suppressor at tyrosine 240. Proc. Natl Acad.
tumor growth. J. Proteome Res. 9(11), receptor tyrosine kinase inhibitor, in patients Sci. USA 109(35), 14164–14169 (2012).
5557–5567 (2010). with recurrent glioblastoma. J. Clin. Oncol. 75 Witusik-Perkowska M, Rieske P, Hulas-
50 Petrik V, Saadoun S, Loosemore A et al. 28(17), 2817–2823 (2010). Bigoszewska K et al. Glioblastoma-derived
Serum alpha 2-HS glycoprotein predicts 63 Ilhan-Mutlu A, Wagner L, Widhalm G et al. spheroid cultures as an experimental model
survival in patients with glioblastoma. Clin. Exploratory investigation of eight circulating for analysis of EGFR anomalies. J.
Chem. 54(4), 713–722 (2008). plasma markers in brain tumor patients. NeuroOncol. 102(3), 395–407 (2011).
51 Popescu ID, Codrici E, Albulescu L et al. Neurosurg. Rev. 36(1), 45–55; discussion 76 Holand K, Salm F, Arcaro A. The
Potential serum biomarkers for glioblastoma 55–6 (2013). phosphoinositide 3-kinase signaling pathway
diagnostic assessedby proteomic approaches. 64 Kumar R, Kamdar D, Madden L et al. Th1/ as a therapeutic target in grade IV brain
Proteome Sci. (In Press). Th2 cytokine imbalance in meningioma, tumors. Curr. Cancer Drug Targets 11(8),
52 Gautam P, Nair SC, Gupta MK et al. Proteins anaplastic astrocytoma and glioblastoma 894–918 (2011).
with altered levels in plasma from multiforme patients. Oncol. Rep. 15(6), 77 Boller D, Doepfner KT, De Laurentiis A et al.
glioblastoma patients as revealed by 1513–1516 (2006). Targeting PI3KC2beta impairs proliferation
iTRAQ-based quantitative proteomic 65 Zagzag D, Salnikow K, Chiriboga L et al. and survival in acute leukemia, brain tumours
analysis. PLoS ONE 7(9), e46153 (2012). Downregulation of major histocompatibility and neuroendocrine tumours. Anticancer Res.
53 Mishra P, Banerjee D, Ben-Baruch A. complex antigens in invading glioma cells: 32(8), 3015–3027 (2012).
Chemokines at the crossroads of tumor- stealth invasion of the brain. Lab. Invest. 78 Opel D, Westhoff MA, Bender A, Braun V,
fibroblast interactions that promote 85(3), 328–341 (2005). Debatin KM, Fulda S. Phosphatidylinositol
malignancy. J. Leukoc. Biol. 89(1), 31–39 66 Demeule M, Currie JC, Bertrand Y et al. 3-kinase inhibition broadly sensitizes
(2011). Involvement of the low-density lipoprotein glioblastoma cells to death receptor- and
54 Calatozzolo C, Canazza A, Pollo B et al. receptor-related protein in the transcytosis of drug-induced apoptosis. Cancer Res. 68(15),
Expression of the new CXCL12 receptor, the brain delivery vector angiopep-2. J. 6271–6280 (2008).
CXCR7, in gliomas. Cancer Biol. Ther. 11(2), NeuroChem. 106(4), 1534–1544 (2008). 79 Kim YW, Liu TJ, Koul D et al. Identification
242–253 (2011). 67 Sun L, Hui AM, Su Q et al. Neuronal and of novel synergistic targets for rational
55 Albulescu A, Codrici E, Popescu ID et al. glioma-derived stem cell factor induces drug combinations with PI3 kinase inhibitors
Cytokine patterns in brain tumour angiogenesis within the brain. Cancer Cell using siRNA synthetic lethality screening
progression. Mediators Inflamm. 2013, 9(4), 287–300 (2006). against GBM. Neuro. Oncol. 13(4), 367–375
979748 (2013).   (2011).
68 Guan M, Jin J, Su B, Liu WW, Lu Y. Tissue
factor expression and angiogenesis in human 80 Sunayama J, Matsuda K, Sato A et al.
• Of interest for advancement in brain tumor
glioma. Clin. BioChem. 35(4), 321–325 Crosstalk between the PI3K/mTOR and
progression
(2002). MEK/ERK pathways involved in the
56 Balkwill F, Mantovani A. Inflammation and maintenance of self-renewal and
cancer: back to Virchow? Lancet 357(9255), 69 Maru SV, Holloway KA, Flynn G et al.
tumorigenicity of glioblastoma stem-like cells.
539–545 (2001). Chemokine production and chemokine
Stem. Cells 28(11), 1930–1939 (2010).
receptor expression by human glioma cells:
57 Vilgrain I, Sidibe A, Polena H et al. Evidence 81 Garlich JR, De P, Dey N et al. A vascular
role of CXCL10 in tumour cell proliferation.
for post-translational processing of vascular targeted pan phosphoinositide 3-kinase
J. Neuroimmunol. 199(1–2), 35–45 (2008).
endothelial (VE)-cadherin in brain tumors:

future science group www.futuremedicine.com 523


Review  Tanase, Albulescu, Codrici et al.

inhibitor prodrug, SF1126, with antitumor development advances and challenges. oncogenesis. In: Stem Cells And Cancer.
and antiangiogenic activity. Cancer Res. 68(1), Pharmacol. Ther. 141(2), 140–149 (2014). Humana Press, MA, USA, 115–26 (2009).
206–215 (2008). 86 Martinho O, Silva-Oliveira R, Miranda- 91 Gatti M, Pattarozzi A, Bajetto A et al.
82 Ylivinkka I, Hu Y, Chen P et al. Netrin-1- Goncalves V et al. In vitro and In vivo analysis Inhibition of CXCL12/CXCR4 autocrine/
induced activation of Notch signaling of RTK inhibitor efficacy and identification of paracrine loop reduces viability of human
mediates glioblastoma cell invasion. J. Cell its novel targets in glioblastomas. Transl. glioblastoma stem-like cells affecting
Sci. 126(Pt 11), 2459–2469 (2013). Oncol. 6(2), 187–196 (2013). self-renewal activity. Toxicology 314(2–3),
83 Tolcher AW, Messersmith WA, Mikulski SM 87 Tănase C, Arsene D, Codorean E et al. 209–220 (2013).
et al. Phase I study of RO4929097, a gamma Elevated expression of cav-1 in brain tumors 92 Tchorz JS, Tome M, Cloetta D et al.
secretase inhibitor of Notch signaling, in and its correlation with angiogenic markers Constitutive Notch2 signaling in neural stem
patients with refractory metastatic or locally – VEGF, bFGF. Virchows Archiv. 452 (2008).   cells promotes tumorigenic features and
advanced solid tumors. J. Clin. Oncol. 30(19), 88 Wu A, Oh S, Wiesner SM et al. Persistence of astroglial lineage entry. Cell Death Dis. 3,
2348–2353 (2012). CD133 + cells in human and mouse glioma e325 (2012).
84 Krop I, Demuth T, Guthrie T et al. Phase I cell lines: detailed characterization of GL261 93 Chiao MT, Cheng WY, Yang YC, Shen CC,
pharmacologic and pharmacodynamic study glioma cells with cancer stem cell-like Ko JL. Suberoylanilide hydroxamic acid
of the gamma secretase (Notch) inhibitor properties. Stem Cells Dev. 17(1), 173–184 (SAHA) causes tumor growth slowdown and
MK-0752 in adult patients with advanced (2008). triggers autophagy in glioblastoma stem cells.
solid tumors. J. Clin. Oncol. 30(19), 89 Driessens G, Beck B, Caauwe A, Simons BD, Autophagy 9(10), 1509–1526 (2013).
2307–2313 (2012). Blanpain C. Defining the mode of tumour 94 Forsyth PA, Krishna N, Lawn S et al.
• Of interest for advancement in molecular growth by clonal analysis. Nature 488(7412), p75 neurotrophin receptor cleavage by
targets as drugs in clinical trials 527–530 (2012). alpha- and gamma-secretases is required for
90 Bengzon J, Englund E, Salford LG et al. neurotrophin mediated proliferation of brain
85 Takebe N, Nguyen D, Yang SX. Targeting
Glioma stem cells in the context of tumor initiating cells. J. Biol. Chem 289(12),
notch signaling pathway in cancer: clinical
8067–8085 (2014).

524 Future Oncol. (2015) 11(3) future science group

You might also like