Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
. 2015 Jul 22;17(10):1322–1332. doi: 10.1093/neuonc/nov119

Intratumoral heterogeneity in glioblastoma: don't forget the peritumoral brain zone

Jean-Michel Lemée 1, Anne Clavreul 1, Philippe Menei 1
PMCID: PMC4578587  PMID: 26203067

Abstract

Glioblastoma (GB) is the most frequent and aggressive primary tumor of the central nervous system. Prognosis remains poor despite ongoing progress. In cases where the gadolinium-enhanced portion of the GB is completely resected, 90% of recurrences occur at the margin of surgical resection in the macroscopically normal peritumoral brain zone (PBZ). Intratumoral heterogeneity in GB is currently a hot topic in neuro-oncology, and the GB PBZ may be involved in this phenomenon. Indeed, this region, which possesses specific properties, has been less studied than the core of the GB tumor. The high rate of local recurrence in the PBZ and the limited success of targeted therapies against GB demonstrate the need for a better understanding of the PBZ. We present here a review of the literature on the GB PBZ, focusing on its radiological, cellular, and molecular characteristics. We discuss how intraoperative analysis of the PBZ is important for the optimization of surgical resection and the development of targeted therapies against GB.

Keywords: glioblastoma, histology, omics, peritumoral brain zone, radiology, targeted therapies


Glioblastoma (GB) is the most frequent and aggressive primary tumor of the central nervous system (CNS). Its incidence varies depending on the country and the population, with 4.96 cases per 100 000 inhabitants per year.1 Despite ongoing progress in the therapeutic management of GB, prognosis remains poor, with an overall survival of 14–15 months after complete surgical resection and adjuvant radiochemotherapy.2 Typically, radiological recurrence occurs 6–7 months after surgery, quickly followed by clinical relapse.

The heterogeneity of GB is a hot topic in neuro-oncology. The interindividual heterogeneity of GB has been well known since the pioneering work of Burger and Kleihues,37 and several studies have even described intratumoral heterogeneity inside GB at the cellular and molecular level.6,8,9 This intratumoral heterogeneity may lead to a different Verhaak subtype in 2 different samples from the same tumor, or it may even give rise to differences at the cellular level as shown by Patel et al (2014).6,1012 GB intratumoral heterogeneity may influence tumor aggressiveness or response to chemotherapies such as temozolomide.8,13

Intratumoral heterogeneity is not only limited to the tumor, it also involves the peritumoral brain zone (PBZ), which possesses specific properties that contribute to GB heterogeneity. Few studies have focused on the PBZ and its microenvironment, despite the fact that 90% of recurrences occur in this area.14 We present here a review of the PBZ of GB, focusing on its radiological, cellular, and molecular characteristics based on current literature and our findings obtained during the Grand Ouest Glioma Project, a translational research project on intratumoral heterogeneity that we recently conducted.12,1524 We also highlight perspectives for future research on the PBZ that may facilitate the development of new targeted therapies against GB and the optimization of surgical resection.

Characteristics of the Glioblastoma Peritumoral Brain Zone

Radiological Aspect

The PBZ is usually defined radiologically as the brain area surrounding the tumor without contrast enhancement in T1 gadolinium-enhanced 3-dimensional (3D) magnetic resonance imaging (MRI). This region is often hyperintense in T2-weighted (especially T2- fluid-attenuated inversion recovery) acquisition, which reflects vasogenic edema in the vicinity of the tumor and suggests tumor infiltration. This definition of the PBZ delimits an area of several centimeters in width around the tumor, which is the site of tumoral invasion and specific molecular, radiological, and cellular alterations that are not limited to the immediate zone surrounding the contrast-enhanced tumor. We chose this radiological definition of the PBZ because GB-cell infiltration may be several centimeters deep,25,26 such that molecular alterations of the parenchyma may be located more than 1 centimeter from the contrast-enhancing lesion.12,15,17,18,23

A lack of contrast enhancement with gadolinium-based contrast agents during MRI of the PBZ does not necessarily indicate the absence of tumor cells in this area because a minimum concentration of tumor cells in the PBZ is necessary to produce detectable alterations on MRI. Various preoperative MRI sequences can be used to search for tumor infiltration in the PBZ. For example, an increase of the apparent diffusion coefficient (ADC) in the PBZ, which measures the magnitude of water molecule diffusion, is correlated with tumor cell infiltration.2730 In dynamic contrast enhancement (DCE) MRI analysis, the volume transfer coefficient ktrans, which is associated with the microvascular permeability of gliomas, is the most sensitive and specific predictor of brain infiltration and histological grading.23,31 Two recent studies also showed that DCE MRI parameters in specific areas of GB, including the PBZ, are correlated with biomarkers of hypoxia and overall patient survival.32,33 Furthermore, the role of diffusion tensor imaging (DTI) was discussed in a recent review, describing an inverse correlation between fractional anisotropy and infiltration of the PBZ.34,35 One study proposed a tumor infiltration index based on the ratio between the expected and the observed fractional anisotropy to investigate tumor-cell infiltration in the PBZ,36 although this index may not be useful for assessing tumor infiltration in patients with peritumoral vasogenic edema.37

Cellular Content

Tumor Cells

The PBZ shares the same macroscopic aspect as normal brain; however, it is well established that GB cell infiltration extends well beyond the radiological limits of the tumor into the macroscopically normal PBZ, suggesting that GB should be considered a systemic brain disease and not a delineated tumor.5,7,38 GB cells invade the brain via the same extracellular routes used by neural cells along existing structures such as blood vessels and white matter tracts.38 GB cells in the PBZ are typically identified by microscopy with hematoxylin-phloxin-saffron (HPS) staining and/or immunohistochemistry using anti-p53 and anti-Ki-67 antibodies. We recently analyzed 28 GB PBZ biopsies stained with HPS, and we found tumor cell infiltration in one third of the samples.23 We also performed DNA index analysis by flow cytometry on 25 of these PBZs. Eight (32%) displayed aneuploid cells, which made up between 3% and 44% of the samples. Interestingly, the PBZ of 4 patients contained some, but not all, of the aneuploid cell populations identified in their corresponding tumor zone, suggesting that not all tumor clones are able to migrate away from the tumor core. Accordingly, the infiltrating GB cells in the PBZ were recently characterized using primary cell cultures and were shown to differ phenotypically from those isolated from the corresponding mass.25,3941 In particular, residual GB cells (ie, those located at the resection margin) proliferate more quickly and are more invasive than GB cells in the tumor center.25,40 Furthermore, residual and central GB cells respond differently to drug and irradiation challenges in vitro.25 Glas et al (2010) and 2 other studies using 5-aminolevulinic acid-assisted surgery found that residual GB cells have a lower capacity for self-renewal than central GB cells.25,39,42 Several genes may distinguish residual GB cells from central GB cells,25,40,41 including genes involved in the stem cell phenotype (CD133, Sox2, nestin, musashi 1), invasion (Galectin-1, Rac1, Rac3, RhoA GTPases, p27, αvβ3 integrin), cell adhesion (CDH20, PCDH19), migration (SNAI2, NANOG, USP6, DISC1), immune or inflammatory responses (TLR4), and blood vessel formation (HEG1, VEGFR2). The identification of these genes has opened new lines of research to improve the understanding of the molecular features promoting the dissemination and progression of GB.

Reactive Astrocytes

Astrocytes are star-shaped glial cells in the CNS with several important functions including synaptic transmission and information processing.43 Many studies have shown that reactive astrocytes are located around GB cells, but their role in GB progression is not completely understood. Reactive astrocytes promote tumor growth and survival44,45 and stimulate the metastasis of breast and lung cancer cells to the brain by secreting various cytokines.45,46 Astrocytes also enhance the invasive potential of GB cells by producing neurotrophic factors such as TGF-α, CXCL12, S1P, and GDNF.47,48 Astrocytes also strongly express IL1β and may be involved in immune reactions against the tumor.49

Inflammatory Cells

Many studies have identified inflammatory cells in the GB tumor core, particularly tumor-associated macrophages (TAMs) and microglia.50,51 More TAMs are present in GB (grade IV) than in grade II or III gliomas, and their numbers are strongly correlated with intratumoral vascular density.52 Although the abundance of TAMs per se is not associated with the differential survival of GB patients, their activation state (M1 or M2) has some prognostic value.5355 Specifically, TAMs that have differentiated into M2 macrophages act as protumoral macrophages and contribute to disease progression. TAMs are now emerging as a promising target for new adjuvant therapies for GB.51,56

Although many studies have focused on the inflammatory cells present in the GB tumor core, only one study (conducted by Parney et al in 2009) has investigated inflammatory cells in the PBZ.57 In this study, macrophage-like cells were the most common infiltrating inflammatory cells in the PBZ, followed by microglia-like cells and lymphocytes. These inflammatory cells were also less common in the PBZ than in the GB tumor core. It is not currently known whether macrophage-like cells present in the PBZ share characteristics with the TAMs found in the GB tumor core.

Other Stromal Cells

We have isolated, from histologically normal GB PBZ, another population of stromal cells, which we named GB-associated stromal cells (GASCs). GASCs are diploid, do not display the genomic alterations typical of GB cells, and share phenotypic and functional properties with cancer-associated fibroblasts (CAFs) that have been described in the stroma of carcinomas. In particular, GASCs express markers associated with CAFs such as alpha smooth-muscle actin (α-SMA), platelet-derived growth factor receptor-beta (PDGFRβ), S100A4/FSP1, and CD146. They have angiogenic properties and tumor-promoting effects on GB cells in vitro and in vivo.15,16 In a recent study, we showed that 2 extratumoral microenvironments are present in GB patients: an extratumoral microenvironment containing GASCs with procarcinogenic properties and another containing GASCs without such properties.16 Interestingly, Roman-Perez et al (2012) also identified 2 different subtypes of extratumoral microenvironment influencing the aggressiveness and outcome of human breast cancers.58 Thus, the subtype of GASCs present after resection may determine the likelihood and timing of GB recurrence. The origin of GASCs is unknown; however, we showed that these cells share several properties with mesenchymal stem cells (MSCs), suggesting that GASCs are derived from these cells.15

Molecular profile

Immunohistochemistry

Various immunohistochemical studies of the PBZ suggest that the peritumoral compartment undergoes substantial modifications regarding vascularization and other biomolecular features. For example, markers of neovascularization (eg, CD105, nestin, phosphorylated extracellular signal-regulated kinases, and c-Jun NH2 terminal kinases [JNKs]) are expressed in the PBZ, irrespective of the presence of tumor cells.5961 In addition, the expression of CD105, JNK, and nestin in the PBZ is associated with poor prognosis.59,60 Jensen et al (2014)32 analyzed molecular markers of hypoxia in the PBZ and found that lower VEGF expression was predictive of longer progression-free survival. The abundance of adenosine A1 receptors and STAT1 expression are also higher in the PBZ than in the GB and normal brain samples.62,63 These modifications may have neuroprotective effects against GB progression. Indeed, STAT1 is a well-known tumor suppressor protein that acts via the JAK-STAT pathway, and gliomas grow faster in adenosine A1 receptor-deficient mice than in control mice.63,64 The immediate vicinity of the GB is also characterized by high levels of copper and zinc,62 but the pathophysiological effect of these changes is unknown.

“Omics”

Molecular biology has made substantial progress in the past few years, with the development and simplification of “omic” analyses including genomic, transcriptomic, and proteomic analyses. Following these analyses, several molecular classification systems for GB were proposed, which have enabled the identification of specific subgroups of GB differing in terms of clinical features, disease progression, and response to adjuvant therapies. The most known is the Verhaak classification system, which is based on a signature of 840 genes that divide GB into proneural, neural, classical, and mesenchymal subtypes.65 More recently, Sturm et al (2012)66 also proposed a clinically relevant molecular classification system based on IDH1 and H3F3A mutations. These omic analyses have been mostly performed on the GB tumor core, although we recently conducted such an analysis on 10 PBZs during the Grand Ouest Glioma Project.23

Genomic analysis of these 10 PBZs found no genomic alterations in the 6 PBZs considered free of tumor infiltration in histopathological analysis, whereas their corresponding tumor zone presented genomic alterations typical of GB including loss/partial loss of chromosome 10, focal deletion of the CDKN2A/B locus, polysomy of chromosome 7, and focal amplification of EGFR. Genomic alterations were present in the 4 PBZs showing tumor cell infiltration in histopathological analysis, comprising some but not all of the alterations found in their corresponding tumor zones. These PBZs contained classical alterations found in GB, like chromosome 7 polysomy, EGFR amplification, and chromosome 10 deletion, although CDKN2A/2B deletion was rare. This result suggests that chromosome 7 and 10 alterations are downstream events in GB tumorigenesis and that some, but not all, tumor clones are able to migrate from the tumor core to the PBZ, as described above. These results are consistent with the recent work of Mangiola et al (2013), who identified cells with an EGFR amplification in the PBZ.67

The transcriptomic and proteomic analyses that we conducted on these 10 PBZs revealed an intratumoral gradient of gene expression from the tumor core to the PBZ.12,17 The transcriptomic analysis also indicated that the PBZ has a proneural or neural profile, regardless of the adjacent GB subtype.12 Gill et al (2014)68 described similar results and demonstrated (with a larger cohort) that the PBZ in the proneural GB subtype strongly express oligodendrocyte progenitor genes, whereas the PBZ in the mesenchymal GB subtype strongly expresses astrocytic and microglial genes.

We were unable to identify a specific molecular signature of the PBZ with “omic” analyses because of interpatient heterogeneity in these 10 PBZ samples. A large cohort of PBZ samples is thus necessary to identify such a molecular signature, but the constitution of this cohort raises the ethical issue of sampling normal brain tissue around the tumor. Furthermore, the identification of such a signature is complicated by the choice of the control brain sample to be used. Samples from epilepsy patients undergoing brain surgery (EB samples) are commonly used as a control in proteomic or transcriptomic studies. However, we recently showed that EB samples display a tumor-like expression pattern and are not a suitable control for studying the proteomic or transcriptomic profile of the PBZ.18 Mangiola et al (2013) studied the gene expression profile of the PBZ using control tissue obtained from patients operated on for deep cavernomas with radiological signs of recent bleeding.67 They identified 57 genes that were significantly differentially expressed between PBZ and control brain samples. Genes associated with growth and proliferation and cell motility/adhesion were overexpressed, whereas those involved in neurogenesis were largely underexpressed in the GB PBZ. However, only 4 PBZ samples were included, and more PBZ samples are required to identify a molecular signature of this area able to predict the clinical outcome of GB patients.

Piwecka et al (2015) recently compared global microRNA (miRNA) expression in adult malignant gliomas (mainly GBs), the tumor margin, and nontumor brain tissues.69 They found that the expression of 97 miRNAs differed significantly between GB and normal brain and that 22 miRNAs were differentially expressed between the border of the tumor and normal brain. Of note, miR-625 was downregulated in the border of the tumor but not in the GB samples, suggesting that some alterations are specific of the PBZ. Several studies have shown that miRNAs play a role in GB growth by influencing stem cell behavior and that these miRNAs are involved in cellular adaptation to metabolic stress in GB.70,71 It will be important to validate PBZ-specific miRNA alterations in order to develop miRNA-based therapies or new diagnostic applications.

The characteristics of the PBZ are summarized in Table 1 and Figure 1.

Table 1.

Summary of the features of the peritumoral brain zone

Approach Technique Characteristics of the PBZ References
Radiology-MRI T1 • No contrast enhancement
T2 FLAIR • Hypersignal
ADC • Intensity of the signal correlated to tumor cell infiltration 2730
DTI • Inverse correlation between the fractional anisotropy and tumoral infiltration of the PBZ 3437
DCE • Volume transfer coefficient (ktrans) is correlated with tumoral infiltration and histological grading 23,31
• Extracellular volume (Ve) and capillary transit time (Tc) are correlated with molecular markers of hypoxia and overall patient survival 32,33
Cell biology Tumor cells • Tumor cells found in the PBZ differ from those in the corresponding tumor mass 25,3941
• Some, but not all tumor clones, migrate away from the tumor core into the PBZ 23
• High proliferation and invasiveness 25,40
• Different response to drug and radiation challenge in vitro 25
• Migrate along the same route of neural stem cells and share numerous traits with them 38
Reactive astrocytes • Promote tumor growth and survival by secreting cytokines 4446
• May be involved in immune reactions against the tumor 49
Inflammatory cells • Macrophage density proportional to histological grade and vascular density 52
• Tumor-associated macrophages and their activation state have prognostic value 5355
GASCs • Phenotype close to CAFs 15,16
• Angiogenic and tumor-promoting properties in vitro and in vivo, depending on the GASC subtype 15,16,24
Molecular biology Immunohistochemistry • Strong expression of adenosine A1 receptor and STAT1, conferring a neuroprotective effect 6264
• High concentrations of copper and zinc 62
• Expression of CD105, ERKs & JNK, associated with poor prognosis 5961
• Expression of VEGF associated with poor progression-free survival 32
Genomics • Presence of some but not all genomic alterations in the nearby tumor zone
• Loss/partial loss of chromosome 10, polysomy of chromosome 7 and focal amplification of EGFR 23
Transcriptomics • PBZ has a proneural or neural profile, irrespective of the adjacent GB subtype 1012
• Interpatient variability, similar to that observed in the corresponding TZ 1012
• Intratumoral gradient of gene expression from the tumor core to the PBZ 1012
• Overexpression of genes associated with growth and proliferation and cell motility/adhesion and underexpression of those involved in neurogenesis 67
• Nonneoplastic cells in the PBZ and the GB subtype may influence the molecular profile of the PBZ 68
• MicroRNA deregulation in GB and adjacent brain 69
Proteomics • Large interpatient variability complicates the identification of protein markers of the PBZ 18

Abbreviations: ADC, apparent diffusion coefficient; CAFs, cancer-associated fibroblasts; DCE, dynamic contrast enhancement; DTI, diffusion tensor imaging; ERKS, extracellular signal-regulated kinases; FLAIR, fluid-attenuated inversion recovery; GASC, glioblastoma-associated stromal cell; GB, glioblastoma; JNK, c-Jun NH2 terminal kinases; PBZ, peritumoral brain zone.

Fig. 1.

Fig. 1.

Graphical representation of the radiological, cellular and molecular characteristics of the glioblastoma peritumoral brain zone (GB PBZ).

Abbreviations: ADC, apparent diffusion coefficient; CAFs, cancer-associated fibroblasts; DTI, diffusion tensor imaging; GASCs, glioblastoma-associated stromal cells; PBZ, peritumoral brain zone; Tc, capillary transit time; TZ, tumor zone; Ve, extracellular volume.

Perspectives on the Glioblastoma Peritumoral Brain Zone

The Glioblastoma Peritumoral Brain Zone: a Potential Target for Glioblastoma Therapy?

Among the many promising preclinical studies, only a few have been successful in human clinical trials. The most recent example is antiangiogenic therapy, which showed no significant effect on overall survival.72 The 2 latest trials on bevacizumab, a humanized monoclonal antibody against VEGF, found that this treatment increased progression-free survival, but not overall survival.73,74 Other GB-targeted therapies focus on the interleukin 13 receptor alpha 2 (IL13Rα2) and EGFR variant III (EGFRvIII).75,76 These targeted therapies showed promising results in preclinical rodent glioma models but demonstrated limited success in human clinical trials due in part to intratumoral heterogeneity in the expression of these targets.

It is important to further characterize the cellular and molecular components of the PBZ to identify new targets and to develop new treatments for GB. Indeed, this region contains the residual disease from which treatment-resistant recurrent disease emerges.

Cuddapah et al (2014)38 indicated in a recent review that invading GB cells retain numerous biological traits inherited from their neural ancestors (eg, Ca2+-AMPA receptors and the Ca2+-activated K+ channel KCa3.1) that should be exploited therapeutically to impair the migration of these cells.38,77,78 Ruiz-Ontanon et al (2013) also found that αVβ3 integrin, low levels of cytoplasmic p27, and their downstream effector proteins Rac and RhoA GTPases are needed for residual tumor cells from the PBZ to acquire an invasive advantage.40 These signaling pathways may provide targets for novel anti-invasive therapies for the treatment of GBs. Glycogen synthase kinase-3 (GSK-3) and STAT-3 are other important molecules that modulate GB invasion and proliferation. Preclinical studies involving the inhibition of these molecules with specific drugs have shown promising results.7981 The miRNA, miR-625, was recently reported to be downregulated in the PBZ but not in the tumor.69 The aberrant downregulation of miR-625 is associated with the local invasion of gastric cancer cells.82 Therefore, this miRNA may be informative for clinical purposes and for diagnostic and potentially therapeutic applications. We identified 2 subtypes of GASCs in the surgical margins of GB patients: one subtype with tumor-promoting and angiogenic properties and the other without these pro-oncogenic properties.16 The procarcinogenic GASC subtype overexpressed several markers including CSPG4/NG2, nestin, and CD146. These molecules may be prognostic biomarkers of GB and/or targets for GB treatment, as described for other cancers.8386 Macrophages detected in the PBZ may also be useful targets. Further investigation is needed to determine their phenotype and function in the PBZ.

Technological approaches should also be developed to target the cellular and molecular components of the PBZ. All attempts to target drugs to the brain are faced with the serious challenge of crossing the blood-brain-barrier (BBB) that separates the blood from the cerebral parenchyma.8789 Several strategies have been developed to bypass the BBB such as specific drug formulations able to cross the BBB,6 local disruption of the BBB through focalized ultrasound,90,91 direct intratumoral drug delivery using convection-enhanced delivery (CED),92,93 and cellular vectors with endogenous tumor-homing activity such as MSCs.94 To target the delivery of diagnostic and therapeutic drugs to the PBZ of GB, Chekhonin et al (2012) produced immunoliposomal nanocontainers based on antibodies against GFAP and the E2 extracellular fragment of connexin 4395 recognizing reactive astrocytes and migrating glioma cells. Our laboratory has developed and patented a novel nanoscale drug encapsulation system, called lipid nanocapsules (LNCs).90 These LNCs have the advantage of being prepared without organic solvent using a low-energy process and are able to encapsulate various compounds including drugs,96,97 radionuclides,98 DNA,99 siRNA,100 and nuclease-resistant locked nucleic acids.101 These LNCs, which showed promising results in glioma models,9698,102 could be used to target the cellular and molecular components of the GB PBZ through the grafting of antibodies onto their surface or the use of MSCs (Figure 2). We recently demonstrated that these cells, which have endogenous tumor-homing activity and are preferentially found throughout the PBZ, can target the delivery of LNCs to the tumor.16,94,103105

Fig. 2.

Fig. 2.

Nano-biotechnological approaches to target the glioblastoma peritumoral brain zone (GB PBZ). Lipid nanocapsules are promising nanoscale systems for delivering therapeutic molecules to the GB PBZ. They can encapsulate many compounds including drugs, radionuclides, DNA, siRNA, and nuclease-resistant locked nucleic acids. Their association with antibodies against molecular components overexpressed in the PBZ such as αVβ3 integrin, KCa3.1, CD146, and CSPG4/NG2 can be used to specifically target the tumor cells and/or GASCs present in the PBZ. Their combination with mesenchymal stem cells, which have endogenous tumor-homing activity and are targeted to the peritumoral region, is another strategy to improve the delivery of therapeutic agents to the PBZ. Abbreviations: Ca-AMPA, Ca2+ permeable AMPA receptor; CSF-1R, colony stimulating factor 1 receptor; CSPG4, chondroitin sulfate proteoglycan; GASC, GB-associated stromal cells; GFAP, glial fibrillary acidic protein; KCa3.1, Ca2+-activated K+ channel.

Glioblastoma Peritumoral Brain Zone: Intraoperative Assessment for Optimal GB Resection?

Clearly, the quality of the surgical resection is critical for the overall survival of the patient and the patient's quality of life and response to adjuvant therapies.106108 The intraoperative assessment of the GB PBZ to detect tumor infiltration is extremely important to optimize surgical resection. The PBZ is typically assessed preoperatively by extemporaneous histopathological examination with a smear test, which provides initial information about the histological nature of the sampled tissue and the presence of tumor cell infiltration. However, this examination lacks sensitivity, and the sample is not representative of the entire resection cavity. Thus, new techniques are required to identify tumor infiltration in the area surrounding the tumor.

Besides examining the PBZ via the preoperative MRI sequences described above, the development of intraoperative MRI in glioma surgery is also a potentially valuable option for optimizing the quality of surgical resection. Intraoperative MRI has shown promising results with a gross total resection rate of 96% in high-grade glioma, which is substantially higher than the classical resection rates of 27%–35% reported in the literature.106,108,109 However, intraoperative MRI has less influence on patient survival, with one controlled study reporting an average intraoperative survival of 226 days in the intraoperative MRI group vs 154 days in the control group.109 Discussion is ongoing to determine whether intraoperative MRI is useful for neurosurgery because similar results in terms of survival can be obtained using alternative techniques (eg, fluorescence-guided surgery with 5-aminolevulinic acid) at a much lower cost.110,111 However, fluorescence-guided surgery is unable to remove the entire tumor because isolated tumor cells may remain outside the fluorescent tumor in the PBZ.39 Thus, spectroscopic devices capable of detecting and quantifying fluorescent signals more precisely than the surgeon's eyes are needed.42,112

A new technique for analyzing the PBZ, optical biopsy, is emerging in different stages of development (Table 2). These optical biopsy techniques are divided into 2 categories: in vivo and ex vivo.

Table 2.

Description of the different techniques available for intraoperative peritumoral brain zone analysis

Technique Type of Technique Spatial Resolution Depth of Penetration Acquisition Surface Time for Image Acquisition & Processing
Optic coherence tomography Ex vivo 1 µm 200 µm Whole brain sample <5 min
Biphotonic microscopy Ex vivo 0.7 µm 400 µm Whole brain sample 5–45 min
Confocal endomicroscopy In vivo 1 µm 250 µm Probe surface Real time
Raman spectrometry In vivo 1 µm 1 µm Probe surface Real time
Intraoperative MRI In vivo <1 mm Whole brain Whole brain 15–20 min

Ex vivo optical biopsies require that brain tissue be sampled during surgery because of technical limitations. However, thanks to recent technological advances, in vivo optical biopsies may hopefully replace ex vivo optical biopsies. Among these techniques, optic coherence tomography imaging uses interference for precise localization of light deep inside tissue. This technique enables noncontact assessment of the brain surface up to 200 µm deep with a spatial resolution of 1 µm, thus allowing discrimination between normal brain, tumor tissue, necrosis, and peripheral infiltrated brain.113,114

Biphotonic microscopy is also currently under evaluation for analysis of the PBZ. This technique allows fast and label-free analysis of brain samples, identifies non-centrosymmetrical structures of the brain (eg, collagen), and provides an overview of the extracellular matrix architecture with a resolution below 1 µm and a depth of 400 µm.115,116

In vivo optical biopsies allow assessment of tumor infiltration during surgery, thus enabling optimal surgical resection. One promising in vivo technique is near-infrared confocal endomicroscopy, which can identify isolated tumor cells in the PBZ during surgery at a precision less than 1 µm and a depth of 250 µm.117,118 This technique, however, is not label-free and requires intravenous injection of dyes such as indocyanine green during surgery. Raman spectroscopy is another in vivo optical biopsy technique based on modification of a laser's wavelength to reflect the brain parenchyma at an axial and depth resolution of 1 µm. This technique has been used in preliminary studies to assess the histological nature of brain samples and to identify tumor remnants in the surgical resection cavity.119121 Karabeber et al (2014) have used gold-silica surface-enhanced Raman scattering nanoparticles and a hand-held Raman scanner to perform image-guided surgical resection of GBs in a transgenic murine model.122

Conclusion

We have much to learn from the PBZ about the recurrence of GB. The PBZ contains specific tumor and stromal cells that promote GB growth and invasion. Given the interpatient heterogeneity of GB PBZ, large-scale studies are required to further characterize this area in order to identify new molecular targets that could be translated into routine clinical practice. Technical progress is also needed to assess the presence of tumor infiltration in the PBZ during surgery and thus optimize the quality of surgical resection. In our opinion, intraoperative MRI is currently the most reliable, easy-to-use technique for analyzing and delineating brain tumors. This recent progress regarding GB and its peritumoral characteristics will facilitate development of personalized targeted therapy and adjuvant treatments of GB after surgery.

Funding

The first author, J.-M.L., has received grants from the Société Française de Neurochirurgie and the Institut National de la Santé et de la Recherche Médicale.

Conflict of interest statement. The authors have no conflict of interest to disclose.

References

  • 1.Loiseau H, Huchet A, Rué M, et al. [Epidemiology of primary brain tumor] [in French]. Rev Neurol (Paris). 2009;165(8–9):650–670. [DOI] [PubMed] [Google Scholar]
  • 2.Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. [DOI] [PubMed] [Google Scholar]
  • 3.Aum DJ, Kim DH, Beaumont TL, et al. Molecular and cellular heterogeneity: the hallmark of glioblastoma. Neurosurg Focus. 2014;37(6):E11. [DOI] [PubMed] [Google Scholar]
  • 4.Friedmann-Morvinski D. Glioblastoma heterogeneity and cancer cell plasticity. Crit Rev Oncog. 2014;19(5):327–336. [DOI] [PubMed] [Google Scholar]
  • 5.Burger PC, Kleihues P. Cytologic composition of the untreated glioblastoma with implications for evaluation of needle biopsies. Cancer. 1989;63(10):2014–2023. [DOI] [PubMed] [Google Scholar]
  • 6.Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344(6190):1396–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Scherer HJ. The forms of growth in gliomas and their practical significance. Brain. 1940;63(1):1–35. [Google Scholar]
  • 8.Meyer M, Reimand J, Lan X, et al. Single cell-derived clonal analysis of human glioblastoma links functional and genomic heterogeneity. Proc Natl Acad Sci USA. 2015;112(3):851–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Soeda A, Hara A, Kunisada T, et al. The evidence of glioblastoma heterogeneity. Sci Rep. 2015;5:7979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Eder K, Kalman B. Molecular heterogeneity of glioblastoma and its clinical relevance. Pathol Oncol Res. 2014;20(4):777–787. [DOI] [PubMed] [Google Scholar]
  • 11.Sottoriva A, Spiteri I, Piccirillo SGM, et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci USA. 2013;110(10):4009–4014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Aubry M, de Tayrac M, Saikali S, et al. “From the core to beyond the margin”: a genomic picture of glioblastoma intratumor heterogeneity. Oncotarget. 2015;6(14):12094–12109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Stieber D, Golebiewska A, Evers L, et al. Glioblastomas are composed of genetically divergent clones with distinct tumourigenic potential and variable stem cell-associated phenotypes. Acta Neuropathol. 2014;127(2):203–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Petrecca K, Guiot M-C, Panet-Raymond V, et al. Failure pattern following complete resection plus radiotherapy and temozolomide is at the resection margin in patients with glioblastoma. J Neurooncol. 2013;111(1):19–23. [DOI] [PubMed] [Google Scholar]
  • 15.Clavreul A, Etcheverry A, Chassevent A, et al. Isolation of a new cell population in the glioblastoma microenvironment. J Neurooncol. 2012;106(3):493–504. [DOI] [PubMed] [Google Scholar]
  • 16.Clavreul A, Guette C, Faguer R, et al. Glioblastoma-associated stromal cells (GASCs) from histologically normal surgical margins have a myofibroblast phenotype and angiogenic properties. J Pathol. 2014;233(1):74–88. [DOI] [PubMed] [Google Scholar]
  • 17.Com E, Clavreul A, Lagarrigue M, et al. Quantitative proteomic Isotope-Coded Protein Label (ICPL) analysis reveals alteration of several functional processes in the glioblastoma. J Proteomics. 2012;75(13):3898–3913. [DOI] [PubMed] [Google Scholar]
  • 18.Lemée J-M, Com E, Clavreul A, et al. Proteomic analysis of glioblastomas: what is the best brain control sample? J Proteomics. 2013;85:165–173. [DOI] [PubMed] [Google Scholar]
  • 19.De Tayrac M, Etcheverry A, Aubry M, et al. Integrative genome-wide analysis reveals a robust genomic glioblastoma signature associated with copy number driving changes in gene expression. Genes Chromosomes Cancer. 2009;48(1):55–68. [DOI] [PubMed] [Google Scholar]
  • 20.De Tayrac M, Aubry M, Saïkali S, et al. A 4-gene signature associated with clinical outcome in high-grade gliomas. Clin Cancer Res. 2011;17(2):317–327. [DOI] [PubMed] [Google Scholar]
  • 21.De Tayrac M, Saikali S, Aubry M, et al. Prognostic significance of EDN/RB, HJURP, p60/CAF-1 and PDLI4, four new markers in high-grade gliomas. PloS One. 2013;8(9):e73332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Etcheverry A, Aubry M, de Tayrac M, et al. DNA methylation in glioblastoma: impact on gene expression and clinical outcome. BMC Genomics. 2010;11:701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lemée J-M, Clavreul A, Aubry M, et al. Characterizing the peritumoral brain zone in glioblastoma: a multidisciplinary analysis. J Neurooncol. 2015;122(1):53–61. [DOI] [PubMed] [Google Scholar]
  • 24.Clavreul A, Etcheverry A, Tétaud C, et al. Identification of two glioblastoma-associated stromal cell subtypes with different carcinogenic properties in histologically normal surgical margins. J Neurooncol. 2015;122(1):1–10. [DOI] [PubMed] [Google Scholar]
  • 25.Glas M, Rath BH, Simon M, et al. Residual tumor cells are unique cellular targets in glioblastoma. Ann Neurol. 2010;68(2):264–269 10.1002/ana.22036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Silbergeld DL, Chicoine MR. Isolation and characterization of human malignant glioma cells from histologically normal brain. J Neurosurg. 1997;86(3):525–531 10.3171/jns.1997.86.3.0525. [DOI] [PubMed] [Google Scholar]
  • 27.Nakai K, Nawashiro H, Shima K, et al. An analysis of T2 mapping on brain tumors. Acta Neurochir Suppl. 2013;118:195–199. [DOI] [PubMed] [Google Scholar]
  • 28.Pauleit D, Langen K-J, Floeth F, et al. Can the apparent diffusion coefficient be used as a noninvasive parameter to distinguish tumor tissue from peritumoral tissue in cerebral gliomas? J Magn Reson Imaging. 2004;20(5):758–764 10.1002/jmri.20177. [DOI] [PubMed] [Google Scholar]
  • 29.Min Z, Niu C, Rana N, et al. Differentiation of pure vasogenic edema and tumor-infiltrated edema in patients with peritumoral edema by analyzing the relationship of axial and radial diffusivities on 3.0 T MRI. Clin Neurol Neurosurg. 2013;115(8):1366–1370. [DOI] [PubMed] [Google Scholar]
  • 30.Wijnen JP, Idema AJS, Stawicki M, et al. Quantitative short echo time 1H MRSI of the peripheral edematous region of human brain tumors in the differentiation between glioblastoma, metastasis, and meningioma. J Magn Reson Imaging. 2012;36(5):1072–1082. [DOI] [PubMed] [Google Scholar]
  • 31.Zhang N, Zhang L, Qiu B, et al. Correlation of volume transfer coefficient Ktrans with histopathologic grade s of gliomas. J Magn Reson Imaging. 2012;36(2):355–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jensen RL, Mumert ML, Gillespie DL, et al. Preoperative dynamic contrast-enhanced MRI correlates with molecular markers of hypoxia and vascularity in specific areas of intratumoral microenvironment and is predictive of patient outcome. Neuro Oncol. 2014;16(2):280–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jalali S, Chung C, Foltz W, et al. MRI biomarkers identify the differential response of glioblastoma multiforme to anti-angiogenic therapy. Neuro Oncol. 2014;16(6):868–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sundgren PC, Fan X, Weybright P, et al. Differentiation of recurrent brain tumor versus radiation injury using diffusion tensor imaging in patients with new contrast-enhancing lesions. Magn Reson Imaging. 2006;24(9):1131–1142. [DOI] [PubMed] [Google Scholar]
  • 35.Deng Z, Yan Y, Zhong D, et al. Quantitative analysis of glioma cell invasion by diffusion tensor imaging. J Clin Neurosci. 2010;17(12):1530–1536. [DOI] [PubMed] [Google Scholar]
  • 36.Lu S, Ahn D, Johnson G, et al. Diffusion-tensor MR imaging of intracranial neoplasia and associated peritumoral edema: introduction of the tumor infiltration index. Radiology. 2004;232(1):221–228. [DOI] [PubMed] [Google Scholar]
  • 37.Kinoshita M, Goto T, Okita Y, et al. Diffusion tensor-based tumor infiltration index cannot discriminate vasogenic edema from tumor-infiltrated edema. J Neurooncol. 2010;96(3):409–415. [DOI] [PubMed] [Google Scholar]
  • 38.Cuddapah VA, Robel S, Watkins S, et al. A neurocentric perspective on glioma invasion. Nat Rev Neurosci. 2014;15(7):455–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Piccirillo SGM, Dietz S, Madhu B, et al. Fluorescence-guided surgical sampling of glioblastoma identifies phenotypically distinct tumour-initiating cell populations in the tumour mass and margin. Br J Cancer. 2012;107(3):462–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ruiz-Ontañon P, Orgaz JL, Aldaz B, et al. Cellular plasticity confers migratory and invasive advantages to a population of glioblastoma-initiating cells that infiltrate peritumoral tissue. Stem Cells. 2013;31(6):1075–1085. [DOI] [PubMed] [Google Scholar]
  • 41.Toussaint LG, Nilson AE, Goble JM, et al. Galectin-1, a gene preferentially expressed at the tumor margin, promotes glioblastoma cell invasion. Mol Cancer. 2012;11:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rampazzo E, Della Puppa A, Frasson C, et al. Phenotypic and functional characterization of Glioblastoma cancer stem cells identified through 5-aminolevulinic acid-assisted surgery. J Neurooncol. 2014;116(3):505–513. [DOI] [PubMed] [Google Scholar]
  • 43.Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119(1):7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lin Q, Balasubramanian K, Fan D, et al. Reactive astrocytes protect melanoma cells from chemotherapy by sequestering intracellular calcium through gap junction communication channels. Neoplasia. 2010;12(9):748–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fitzgerald DP, Palmieri D, Hua E, et al. Reactive glia are recruited by highly proliferative brain metastases of breast cancer and promote tumor cell colonization. Clin Exp Metastasis. 2008;25(7):799–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Seike T, Fujita K, Yamakawa Y, et al. Interaction between lung cancer cells and astrocytes via specific inflammatory cytokines in the microenvironment of brain metastasis. Clin Exp Metastasis. 2011;28(1):13–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rath BH, Fair JM, Jamal M, et al. Astrocytes enhance the invasion potential of glioblastoma stem-like cells. PloS One. 2013;8(1):e54752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kim J-K, Jin X, Sohn Y-W, et al. Tumoral RANKL activates astrocytes that promote glioma cell invasion through cytokine signaling. Cancer Lett. 2014;353(2):194–200. [DOI] [PubMed] [Google Scholar]
  • 49.Nagashima G, Suzuki R, Asai J, et al. Immunohistochemical analysis of reactive astrocytes around glioblastoma: an immunohistochemical study of postmortem glioblastoma cases. Clin Neurol Neurosurg. 2002;104(2):125–131. [DOI] [PubMed] [Google Scholar]
  • 50.Charles NA, Holland EC, Gilbertson R, et al. The brain tumor microenvironment. Glia. 2012;60(3):502–514. [DOI] [PubMed] [Google Scholar]
  • 51.Glass R, Synowitz M. CNS macrophages and peripheral myeloid cells in brain tumours. Acta Neuropathol. 2014;128(3):347–362. [DOI] [PubMed] [Google Scholar]
  • 52.Nishie A, Ono M, Shono T, et al. Macrophage infiltration and heme oxygenase-1 expression correlate with angiogenesis in human gliomas. Clin Cancer Res. 1999;5(5):1107–1113. [PubMed] [Google Scholar]
  • 53.Komohara Y, Ohnishi K, Kuratsu J, et al. Possible involvement of the M2 anti-inflammatory macrophage phenotype in growth of human gliomas. J Pathol. 2008;216(1):15–24. [DOI] [PubMed] [Google Scholar]
  • 54.Engler JR, Robinson AE, Smirnov I, et al. Increased microglia/macrophage gene expression in a subset of adult and pediatric astrocytomas. PloS One. 2012;7(8):e43339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Pyonteck SM, Akkari L, Schuhmacher AJ, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19(10):1264–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Bowman RL, Joyce JA. Therapeutic targeting of tumor-associated macrophages and microglia in glioblastoma. Immunotherapy. 2014;6(6):663–666. [DOI] [PubMed] [Google Scholar]
  • 57.Parney IF, Waldron JS, Parsa AT. Flow cytometry and in vitro analysis of human glioma-associated macrophages. Laboratory investigation. J Neurosurg. 2009;110(3):572–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Román-Pérez E, Casbas-Hernández P, Pirone JR, et al. Gene expression in extratumoral microenvironment predicts clinical outcome in breast cancer patients. Breast Cancer Res. 2012;14(2):R51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mangiola A, Lama G, Giannitelli C, et al. Stem cell marker nestin and c-Jun NH2-terminal kinases in tumor and peritumor areas of glioblastoma multiforme: possible prognostic implications. Clin Cancer Res. 2007;13(23):6970–6977. [DOI] [PubMed] [Google Scholar]
  • 60.Sica G, Lama G, Anile C, et al. Assessment of angiogenesis by CD105 and nestin expression in peritumor tissue of glioblastoma. Int J Oncol. 2011;38(1):41–49. [PubMed] [Google Scholar]
  • 61.Lama G, Mangiola A, Anile C, et al. Activated ERK1/2 expression in glioblastoma multiforme and in peritumor tissue. Int J Oncol. 2007;30(6):1333–1342. [PubMed] [Google Scholar]
  • 62.Dehnhardt M, Zoriy MV, Khan Z, et al. Element distribution is altered in a zone surrounding human glioblastoma multiforme. J Trace Elem Med Biol. 2008;22(1):17–23. [DOI] [PubMed] [Google Scholar]
  • 63.Haybaeck J, Obrist P, Schindler CU, et al. STAT-1 expression in human glioblastoma and peritumoral tissue. Anticancer Res. 2007;27(6B):3829–3835. [PubMed] [Google Scholar]
  • 64.Synowitz M, Glass R, Färber K, et al. A1 adenosine receptors in microglia control glioblastoma-host interaction. Cancer Res. 2006;66(17):8550–8557. [DOI] [PubMed] [Google Scholar]
  • 65.Verhaak RGW, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17(1):98–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Sturm D, Witt H, Hovestadt V, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012;22(4):425–437. [DOI] [PubMed] [Google Scholar]
  • 67.Mangiola A, Saulnier N, De Bonis P, et al. Gene expression profile of glioblastoma peritumoral tissue: an ex vivo study. PloS One. 2013;8(3):e57145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gill BJ, Pisapia DJ, Malone HR, et al. MRI-localized biopsies reveal subtype-specific differences in molecular and cellular composition at the margins of glioblastoma. Proc Natl Acad Sci USA. 2014;111(34):12550–12555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Piwecka M, Rolle K, Belter A, et al. Comprehensive analysis of microRNA expression profile in malignant glioma tissues. Mol Oncol. 2015;pii:S1574–7891(15)00056-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Godlewski J, Nowicki MO, Bronisz A, et al. MicroRNA-451 regulates LKB1/AMPK signaling and allows adaptation to metabolic stress in glioma cells. Mol Cell. 2010;37(5):620–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Godlewski J, Newton HB, Chiocca EA, et al. MicroRNAs and glioblastoma; the stem cell connection. Cell Death Differ. 2010;17(2):221–228. [DOI] [PubMed] [Google Scholar]
  • 72.Bastien JIL, McNeill KA, Fine HA. Molecular characterizations of glioblastoma, targeted therapy, and clinical results to date. Cancer. 2015;121(4):502–516. [DOI] [PubMed] [Google Scholar]
  • 73.Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):709–722. [DOI] [PubMed] [Google Scholar]
  • 74.Gilbert MR, Sulman EP, Mehta MP. Bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370(21):2048–2049. [DOI] [PubMed] [Google Scholar]
  • 75.Thaci B, Brown CE, Binello E, et al. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro Oncol. 2014;16(10):1304–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Addeo R, Zappavigna S, Parlato C, et al. Erlotinib: early clinical development in brain cancer. Expert Opin Investig Drugs. 2014;23(7):1027–1037. [DOI] [PubMed] [Google Scholar]
  • 77.Cuddapah VA, Turner KL, Seifert S, et al. Bradykinin-induced chemotaxis of human gliomas requires the activation of KCa3.1 and ClC-3. J Neurosci. 2013;33(4):1427–1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ishiuchi S, Tsuzuki K, Yoshida Y, et al. Blockage of Ca(2+)-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat Med. 2002;8(9):971–978. [DOI] [PubMed] [Google Scholar]
  • 79.Nowicki MO, Dmitrieva N, Stein AM, et al. Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro Oncol. 2008;10(5):690–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Williams SP, Nowicki MO, Liu F, et al. Indirubins decrease glioma invasion by blocking migratory phenotypes in both the tumor and stromal endothelial cell compartments. Cancer Res. 2011;71(16):5374–5380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Agudelo-Garcia PA, De Jesus JK, Williams SP, et al. Glioma cell migration on three-dimensional nanofiber scaffolds is regulated by substrate topography and abolished by inhibition of STAT3 signaling. Neoplasia. 2011;13(9):831–840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wang M, Li C, Nie H, et al. Down-regulated miR-625 suppresses invasion and metastasis of gastric cancer by targeting ILK. FEBS Lett. 2012;586(16):2382–2388. [DOI] [PubMed] [Google Scholar]
  • 83.Svendsen A, Verhoeff JJC, Immervoll H, et al. Expression of the progenitor marker NG2/CSPG4 predicts poor survival and resistance to ionising radiation in glioblastoma. Acta Neuropathol. 2011;122(4):495–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Chekenya M, Enger PØ, Thorsen F, et al. The glial precursor proteoglycan, NG2, is expressed on tumour neovasculature by vascular pericytes in human malignant brain tumours. Neuropathol Appl Neurobiol. 2002;28(5):367–380. [DOI] [PubMed] [Google Scholar]
  • 85.Hatanpaa KJ, Hu T, Vemireddy V, et al. High expression of the stem cell marker nestin is an adverse prognostic factor in WHO grade II-III astrocytomas and oligoastrocytomas. J Neurooncol. 2014;117(1):183–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Akiyama M, Matsuda Y, Ishiwata T, et al. Inhibition of the stem cell marker nestin reduces tumor growth and invasion of malignant melanoma. J Invest Dermatol. 2013;133(5):1384–1387. [DOI] [PubMed] [Google Scholar]
  • 87.Stockwell J, Abdi N, Lu X, et al. Novel central nervous system drug delivery systems. Chem Biol Drug Des. 2014;83(5):507–520. [DOI] [PubMed] [Google Scholar]
  • 88.Tajes M, Ramos-Fernández E, Weng-Jiang X, et al. The blood-brain barrier: structure, function and therapeutic approaches to cross it. Mol Membr Biol. 2014;31(5):152–167. [DOI] [PubMed] [Google Scholar]
  • 89.Bhowmik A, Khan R, Ghosh MK. Blood brain barrier: a challenge for effectual therapy of brain tumors. BioMed Res Int. 2015;2015:320941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Huynh NT, Passirani C, Saulnier P, et al. Lipid nanocapsules: a new platform for nanomedicine. Int J Pharm. 2009;379(2):201–209. [DOI] [PubMed] [Google Scholar]
  • 91.Burgess A, Hynynen K. Drug delivery across the blood-brain barrier using focused ultrasound. Expert Opin Drug Deliv. 2014;11(5):711–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lonser RR, Sarntinoranont M, Morrison PF, et al. Convection-enhanced delivery to the central nervous system. J Neurosurg. 2015;122(3):697–706. [DOI] [PubMed] [Google Scholar]
  • 93.Vogelbaum MA, Aghi MK. Convection-enhanced delivery for the treatment of glioblastoma. Neuro Oncol. 2015;17(Suppl 2):ii3–ii8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Roger M, Clavreul A, Venier-Julienne M-C, et al. The potential of combinations of drug-loaded nanoparticle systems and adult stem cells for glioma therapy. Biomaterials. 2011;32(8):2106–2116. [DOI] [PubMed] [Google Scholar]
  • 95.Chekhonin VP, Baklaushev VP, Yusubalieva GM, et al. Targeted delivery of liposomal nanocontainers to the peritumoral zone of glioma by means of monoclonal antibodies against GFAP and the extracellular loop of Cx43. Nanomedicine. 2012;8(1):63–70. [DOI] [PubMed] [Google Scholar]
  • 96.Allard E, Huynh NT, Vessières A, et al. Dose effect activity of ferrocifen-loaded lipid nanocapsules on a 9L-glioma model. Int J Pharm. 2009;379(2):317–323. [DOI] [PubMed] [Google Scholar]
  • 97.Laine A-L, Huynh NT, Clavreul A, et al. Brain tumour targeting strategies via coated ferrociphenol lipid nanocapsules. Eur J Pharm Biopharm. 2012;81(3):690–693. [DOI] [PubMed] [Google Scholar]
  • 98.Vanpouille-Box C, Lacoeuille F, Belloche C, et al. Tumor eradication in rat glioma and bypass of immunosuppressive barriers using internal radiation with (188)Re-lipid nanocapsules. Biomaterials. 2011;32(28):6781–6790. [DOI] [PubMed] [Google Scholar]
  • 99.David S, Montier T, Carmoy N, et al. Treatment efficacy of DNA lipid nanocapsules and DNA multimodular systems after systemic administration in a human glioma model. J Gene Med. 2012;14(12):769–775. [DOI] [PubMed] [Google Scholar]
  • 100.Resnier P, David S, Lautram N, et al. EGFR siRNA lipid nanocapsules efficiently transfect glioma cells in vitro. Int J Pharm. 2013;454(2):748–755. [DOI] [PubMed] [Google Scholar]
  • 101.Griveau A, Bejaud J, Anthiya S, et al. Silencing of miR-21 by locked nucleic acid-lipid nanocapsule complexes sensitize human glioblastoma cells to radiation-induced cell death. Int J Pharm. 2013;454(2):765–774. [DOI] [PubMed] [Google Scholar]
  • 102.Vinchon-Petit S, Jarnet D, Michalak S, et al. Local implantation of doxorubicin drug eluting beads in rat glioma. Int J Pharm. 2010;402(1–2):184–189. [DOI] [PubMed] [Google Scholar]
  • 103.Roger M, Clavreul A, Venier-Julienne M-C, et al. Mesenchymal stem cells as cellular vehicles for delivery of nanoparticles to brain tumors. Biomaterials. 2010;31(32):8393–8401. [DOI] [PubMed] [Google Scholar]
  • 104.Roger M, Clavreul A, Sindji L, et al. In vitro and in vivo interactions between glioma and marrow-isolated adult multilineage inducible (MIAMI) cells. Brain Res. 2012;1473:193–203. [DOI] [PubMed] [Google Scholar]
  • 105.Roger M, Clavreul A, Huynh NT, et al. Ferrociphenol lipid nanocapsule delivery by mesenchymal stromal cells in brain tumor therapy. Int J Pharm. 2012;423(1):63–68. [DOI] [PubMed] [Google Scholar]
  • 106.Orringer D, Lau D, Khatri S, et al. Extent of resection in patients with glioblastoma: limiting factors, perception of resectability, and effect on survival. J Neurosurg. 2012;117(5):851–859. [DOI] [PubMed] [Google Scholar]
  • 107.Chaichana KL, Jusue-Torres I, Navarro-Ramirez R, et al. Establishing percent resection and residual volume thresholds affecting survival and recurrence for patients with newly diagnosed intracranial glioblastoma. Neuro Oncol. 2014;16(1):113–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.McGirt MJ, Chaichana KL, Gathinji M, et al. Independent association of extent of resection with survival in patients with malignant brain astrocytoma. J Neurosurg. 2009;110(1):156–162. [DOI] [PubMed] [Google Scholar]
  • 109.Senft C, Bink A, Franz K, et al. Intraoperative MRI guidance and extent of resection in glioma surgery: a randomised, controlled trial. Lancet Oncol. 2011;12(11):997–1003. [DOI] [PubMed] [Google Scholar]
  • 110.Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7(5):392–401. [DOI] [PubMed] [Google Scholar]
  • 111.Barone DG, Lawrie TA, Hart MG. Image guided surgery for the resection of brain tumours. Cochrane Database Syst Rev. 2014;1:CD009685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Valdés PA, Kim A, Brantsch M, et al. δ-aminolevulinic acid-induced protoporphyrin IX concentration correlates with histopathologic markers of malignancy in human gliomas: the need for quantitative fluorescence-guided resection to identify regions of increasing malignancy. Neuro Oncol. 2011;13(8):846–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Assayag O, Grieve K, Devaux B, et al. Imaging of non-tumorous and tumorous human brain tissues with full-field optical coherence tomography. NeuroImage Clin. 2013;2:549–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Chen Y, Aguirre AD, Ruvinskaya L, et al. Optical coherence tomography (OCT) reveals depth-resolved dynamics during functional brain activation. J Neurosci Methods. 2009;178(1):162–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Debarbieux F. [Towards a dynamic anatomopathology of glioblastoma in mice thanks to in vivo biphotonic microscopy]. Ann Pathol. 2010;30(5 Suppl 1):53–55. [DOI] [PubMed] [Google Scholar]
  • 116.Lemée J, Clavreul A, Rakotoarimalala S, et al. P16.20 analysis of glioblastoma's peritumoral brain zone using biphotonic microscopy. Neuro Oncol. 2014;16(suppl 2):ii82–ii83. [Google Scholar]
  • 117.Martirosyan NL, Cavalcanti DD, Eschbacher JM, et al. Use of in vivo near-infrared laser confocal endomicroscopy with indocyanine green to detect the boundary of infiltrative tumor. J Neurosurg. 2011;115(6):1131–1138. [DOI] [PubMed] [Google Scholar]
  • 118.Böhringer HJ, Lankenau E, Stellmacher F, et al. Imaging of human brain tumor tissue by near-infrared laser coherence tomography. Acta Neurochir (Wien). 2009;151(5):507–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kalkanis SN, Kast RE, Rosenblum ML, et al. Raman spectroscopy to distinguish grey matter, necrosis, and glioblastoma multiforme in frozen tissue sections. J Neurooncol. 2014;116(3):477–485. [DOI] [PubMed] [Google Scholar]
  • 120.Rutka JT, Kim B, Etame A, et al. Nanosurgical resection of malignant brain tumors: beyond the cutting edge. ACS Nano. 2014;810:9716–9722. [DOI] [PubMed] [Google Scholar]
  • 121.Tanahashi K, Natsume A, Ohka F, et al. Assessment of tumor cells in a mouse model of diffuse infiltrative glioma by Raman spectroscopy. BioMed Res Int. 2014;2014:860241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Karabeber H, Huang R, Iacono P, et al. Guiding brain tumor resection using surface-enhanced Raman scattering nanoparticles and a hand-held Raman scanner. ACS Nano. 2014;810:9755–9766. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press

RESOURCES