Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
. 2014 Sep 24;17(3):343–360. doi: 10.1093/neuonc/nou207

Circulating glioma biomarkers

Johan M Kros 1, Dana M Mustafa 1, Lennard JM Dekker 1, Peter AE Sillevis Smitt 1, Theo M Luider 1, Ping-Pin Zheng 1
PMCID: PMC4483097  PMID: 25253418

Abstract

Validated biomarkers for patients suffering from gliomas are urgently needed for standardizing measurements of the effects of treatment in daily clinical practice and trials. Circulating body fluids offer easily accessible sources for such markers. This review highlights various categories of tumor-associated circulating biomarkers identified in blood and cerebrospinal fluid of glioma patients, including circulating tumor cells, exosomes, nucleic acids, proteins, and oncometabolites. The validation and potential clinical utility of these biomarkers is briefly discussed. Although many candidate circulating protein biomarkers were reported, none of these have reached the required validation to be introduced for clinical practice. Recent developments in tracing circulating tumor cells and their derivatives as exosomes and circulating nuclear acids may become more successful in providing useful biomarkers. It is to be expected that current technical developments will contribute to the finding and validation of circulating biomarkers.

Keywords: biomarker, blood, cerebrospinal fluid, circulating tumor cell, nucleic acid, exosome, glioma, Omics


Gliomas are the most common type of primary brain tumor and have an invariable fatal outcome and dismal prognosis. Each year, ∼200 000 patients are diagnosed with a glioma worldwide.1 Gliomas are subdivided into astrocytoma, oligodendroglioma, and oligoastrocytoma based on immunophenotypical similarity to a cell of putative origin. The tumors are assigned malignancy grades according to WHO criteria.2 Gliomas usually recur and tend to increase in malignancy grade over time (Fig. 1). Glioma progression is accompanied by extensive neovascularization, and the newly formed blood vessels are leaky, which is reflected by tumor enhancement on MRI. Glioblastomas (GBMs) represent astrocytomas of the highest malignancy grade (WHO grade IV) and are the most common gliomas and the most aggressive primary brain tumors in adults, with a median survival of only 14.6 months.3,4 Various molecular aberrations of gliomas (eg, the combined loss of chromosome arms 1p and 19q, the presence of isocitrate dehydrogenase 1(IDH1) mutation, epidermal growth factor receptor (EGFR) amplification, copy number aberrations of chromosomes 7 and 10, and MGMT promoter hypermethylation) harbor diagnostic, prognostic, or predictive information.58 The molecular tests are carried out on tumor biopsies or resection specimens. Mutant IDH1, MGMT promoter methylation, and loss of 1p and 19q can also be detected in serum and cerebrospinal fluid (CSF) of glioma patients, and efforts to trace these aberrations in circulating tumor cells or circulating DNA are ongoing.913

Fig. 1.

Fig. 1.

Astrocytoma in low-grade (A) and high-grade (B) stages. (A) Low-grade astrocytoma (H&E; 200 x; bar = 100 micron). The infiltrating low-grade astrocytoma is composed of genetically altered glial tumor cells that infiltrate into brain tissue and are surrounded by reactive astrocytes (arrows), oligodendrocytes, and microglial cells. The tumor vasculature at this stage is inconspicuous and will be recruited for sprouting angiogenesis. (B) High-grade astrocytoma (grade IV or glioblastoma) (H&E, 200x). The advanced tumor grade is reflected by high cell density, polymorphism of the nuclei of the tumor cells, proliferated blood vessels (“microvascular proliferation”; MVP) and necrosis (NEC). The proliferated blood vessel walls are associated with breakdown of the blood-brain barrier causing leaking vessels, which is represented by enhancement on CT and MRI.

Therapeutic modalities for gliomas include surgical resection, radiotherapy, and chemotherapy. The gold standard for measuring the effects of treatment in patients with gliomas is the application of the Response Assessment in Neuro-Oncology (RANO) criteria to the radiological appearance of the tumor on MRI.14,15 There is considerable variation in the radiological presentation of gliomas and their recurrences.16 A notorious problem in measuring the effects of treatment is so-called pseudoprogression, a treatment-related response of brain tissue to chemotherapy and radiation. Glioma pseudoprogression causes an increase in enhancement and edema on MRI that mimics true tumor progression.17,18 This condition is probably induced by treatment-related local inflammation, resulting in edema and increased abnormal vessel permeability. There is a need for diagnostic discrimination because combined chemotherapy and radiation (the standard of care for GBM) may induce pseudoprogression in ∼30% of cases.19,20 Unfortunately, there are no reliable radiological techniques to distinguish between pseudoprogression and tumor recurrence or progression.21,22 The identification of proliferating tumor cells in tissue biopsies taken in situations of pseudoprogression may be troublesome, and the significance of the presence of scattered cells with morphological or molecular characteristics of the original lesion is disputable (Fig. 2). Currently, there are no biomarkers or radiographic or clinical modalities to reliably distinguish glioma recurrence from radiation necrosis or to monitor tumor response to therapy. Objective measurable parameters for the presence of tumor, tumor activity, and response to treatment would be a welcome addition to the currently available diagnostic arsenal.

Fig. 2.

Fig. 2.

Histology of a glial tumor with effects of radiation therapy. (H&E; 200 x; bar = 100 micron). The radiation therapy has caused homogenization of vascular walls (BV), edema of the neuropil, and proliferation of reactive astrocytes. The glial tumor cells cannot reliably be distinguished from reactive astrocytes because nuclear polymorphism (circles) may be present in both.

Recently, advances in “omics” based technologies, including genomics, transcriptomics, proteomics, and metabolomics, have led to an explosion of activity in the field of biomarker research, particularly related to cancer. The general definition of a biomarker is “a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention”.23,24 Cancer biomarkers include a broad range and level of biochemical entities such as nucleic acids, proteins, sugars, lipids, and small metabolites, and cytogenetic and cytokinetic parameters as well as whole tumor cells and exosomes (microvesicles). The advantage of biomarkers present in blood or CSF is their relatively easy accessibility, which facilitates repetitive measurements with obviously better monitoring of disease. In order to evaluate and compare tumor biomarkers, the “Tumor Marker Utility Grading System” has been proposed by the National Comprehensive Cancer Network (NCCN).25,26 In this system, potential tumor markers are evaluated for their diagnostic, prognostic, or predictive performance as reflected by overall survival, disease-free survival, quality of life, or cost of care.2527 In the NCCN system, levels of evidence have been applied to several potential glioma biomarkers including IDH1 mutation, MGMT methylation, loss of 1p/19q, BRAF fusion, and CpG island methylator phenotype (CIMP).26,27 Among these biomarkers, only 1p/19q testing has been credited for the highest level of evidence because of its ability to improve clinical decision-making and predict patient outcome (IA level).26,27

Because of its anatomical proximity to the CNS, CSF is a promising source of biomarker discovery for diseases of the CNS. CSF samples are used for traditional cytology and have also been recently used for detecting brain metastasis by employing sensitive techniques such as flow cytometry, fluorescence in-situ hybridization (FISH), and PCR/reverse transcription PCR.28,29 The relative low protein concentration of CSF (100–400 times lower as compared with serum) allows rapid screening, low sample consumption, and accurate identification or profiling by proteomic technologies, which have been facilitated by the recent publication of the normal human CSF proteome.28,29 Under pathological conditions, one may find altered levels of normal constitutive proteins or proteins that are usually absent from normal CSF. These proteins may have entered the CSF due to disruption of the blood–brain barrier or intrathecal secretion, or shedding by tumor cells of primary brain tumor or metastasis, and/or their microenvironment. By now, various candidate protein biomarkers for gliomas have been found in CSF.3051

In this review, different categories of tumor-associated circulating biomarkers, which have been identified in blood and CSF of glioma patients, are addressed including circulating tumor cells (CTCs), exosomes (microvesicles), circulating nucleic acids (DNA, RNA and miRNA), proteins, and metabolites.

Circulating Tumor Cells

CTCs are detected when diagnosing metastatic disease or tumor recurrence and may be used to monitor disease progression and therapeutic response. CTCs are found in the peripheral blood of patients with advanced stages of solid cancers with or without clinically detectable metastasis.5256 It has been shown that the presence of CTCs is related to tumor response, progression-free survival, and overall survival in patients suffering from various tumor types and that the presence of CTCs may hint at the existence of a hitherto undiscovered primary tumor.5759 Only one cell per 109 cells represents a CTC in the blood of patients with metastatic cancer, and the specificity and sensitivity of CTC detection is a technical challenge.60 Various detection technologies have been recently developed including microchips, filtration devices, quantitative reverse-transcription PCR assays, automated microscopy systems, and telomerase promoter-based assays.6164 CTCs are particularly valuable for tumor characterization in situations in which tissue biopsies are unavailable or the collected tissue is of poor quality and/or insufficient quantity.63 To what extent CTCs represent the cell population in the solid tumor part remains questionable. Because CTCs reflect the molecular heterogeneity of the tumor, they are important for therapeutic strategies. CTCs are probably not only released from the primary tumor but also from metastatic sites. However, most cancer cells are rapidly destroyed in the circulation, and the metastatic potential of CTCs seems limited.6567 Current clinical investigations of CTCs focus on their molecular characterization and the classification of heterogeneous subsets in relation to treatment resistance. CTCs are subjects of investigations in basal processes of epithelial-mesenchymal transition (EMT), collective cell migration and more, with the aim being to better understand the mechanisms of tumorigenesis, invasion, and metastasis.63,64,68,69

Until recently, the spread of glial tumor cells outside the brain was considered to be a rare event. However, many isolated cases of metastasizing glial neoplasms have been reported.7081 Several cases of transmission of metastatic GBM from donor to organ transplant recipients further supports the notion that appearance of glioma cells in the circulation is not as rare as previously believed and that occurrence rates may match those of other solid tumors.8289 Novel sensitive imaging techniques contribute to higher detection rates. So far, data on circulating CTCs associated with brain tumors are limited, and the use of CTCs as biomarkers in glioma patients is just beginning. The identification of CTCs was carried out by using markers for neural lineage in one study,90 and a telomerase promoter-based assay was used for the detection of these cells in another study (Table 1).64 There are limitations in the use of lineage markers for the identification of CTCs because of overlap in marker expression of tumor cells and normal cells. Further characterization of CTCs at the DNA, RNA, or protein level will improve the identification of true CTCs and their subfractions from other circulating cells.

Table 1.

Circulating tumor cells, tumor-associated nucleic acids, and exosomes in glioma patients

Reference Biomarker Source Time of Take Methodology n Tumor Type Treatment Response Survival Radiology Median Follow-up
Macarthur et al 201464 CTCs Blood Pretreatment TPBA 5 HGG NA NA NA NA
Tumilson et al 2014106 miR-15b CSF NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-15b Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-17–5p Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-20a Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-21 CSF NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-21 Plasma NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-23a Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-31 Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-106a Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-146b Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-148a Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-150 Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-193a Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-197 Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-200b Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-200 CSF NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-221 Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-222 Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-342–3p Plasma NA qRT-PCR NA Glioma NOS NA NA NA NA
miR-548b-5p Serum NA qRT-PCR NA Glioma NOS NA NA NA NA
Salkeni et al 2013109 EGFRvIII Plasma Pre/postoperative PCR 13 GBM Yes NA NA NA
Majchrzak-Celinska et al 201313 Methyl. MGMT Serum Pretreatment PCR 17 AII/GBM NA NA NA NA
Methyl. RASSF1A Serum Pretreatment PCR 17 AII/GBM NA NA NA NA
Methyl. p15INK4B Serum Pretreatment PCR 17 AII/GBM NA NA NA NA
Methyl. p14ARF Serum Pretreatment PCR 17 AII/GBM NA NA NA NA
Chen et al 20139 Mutant IDH1 Serum MVs Intraoperative BEAMng/ddPCR 24 AII/GBM NA NA NA NA
Mutant IDH1 CSF MVs Intraoperative BEAMng/ddPCR 24 AII/GBM NA NA NA NA
Yang et al 2013131 miR-15b Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
miR-23a Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
miR-133a Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
miR-150 Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
miR-197 Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
miR-497 Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
miR-548b-5p Serum Preoperative Sequencing/qRT-PCR 177 AIII NA NA NA NA
Cohen et al 201290 CTCs Blood Pretreatment Cell lineage markers 11 GBM NA NA NA NA
Boisselier et al 2012108 Mutant IDH1 Plasma Posttreatment PCR 80 Astro II-IV NA NA NA NA
Noerholm et al 2012116 Expression patterns Serum MVs Pretreatment Microarray/qRT-PCR 9 GBM NA NA NA NA
Teplyuk et al 2012130 miR-10b CSF Intraop./posttreat. RT-PCR 19 GBM NA NA NA NA
miR-21 CSF Intraop./posttreat. RT-PCR 19 GBM NA NA NA NA
Baraniskin et al 2012132 miR-15b CSF Intraop./postoperative qRT-PCR 10 AII/GBM NA NA NA NA
miR-21 CSF Intraop./postoperative qRT-PCR 10 AII/AIII/GBM NA NA NA NA
Ilhan-Mutlu et al 2012134 miR-21 Plasma Pretreatment qRT-PCR 10 GBM NA NA NA NA
Wang et al 2012135 miR-21 Plasma Pre/posttreatment qRT-PCR 30 GBM Yes NA NA NA
miR-128 Plasma Pre/posttreatment qRT-PCR 30 GBM Yes NA NA NA
miR-342-3p Plasma Pre/posttreatment qRT-PCR 30 GBM Yes NA NA NA
Balana et al 201111 Methyl. MGMT Serum Intraoperative PCR 37 GBM NA Yes NA 120 weeks
Nilsson et al 2011118 EGFRvIII Platelets NA RT-PCR 26 AII/AIII/GBM NA NA NA NA
Roth et al 2011136 miR-128 Blood cell pellets Postoperative qRT-PCR 20 GBM NA NA NA NA
miR-342-3p Blood cell pellets Postoperative qRT-PCR 20 GBM NA NA NA NA
Lavon et al 201012 1p LOH Serum Posttreatment PCR 70 AII/OII/OIII NA NA NA NA
10q LOH Serum Posttreatment PCR 70 AII/OII/OIII NA NA NA NA
19q LOH Serum Posttreatment PCR 70 AII/OII/OIII NA NA NA NA
Methyl. MGMT Serum Posttreatment PCR 70 AII/OII/OIII NA NA NA NA
Methyl. PTEN Serum Posttreatment PCR 70 AII/OII/OIII NA NA NA NA
Liu et al 201010 Methyl. MGMT Serum Pretreatment MeDIP/PCR 66 AOA/GBM NA Yes NA 11.3 months
Methyl. p16 Serum Pretreatment MeDIP/PCR 66 AOA/GBM NA Yes NA 11.3 months
Methyl. TIMP-3 Serum Pretreatment MeDIP/PCR 66 AOA/GBM NA No NA 11.3 months
Methyl. THBS1 Serum Pretreatment MeDIP/PCR 66 AOA/GBM NA Yes NA 11.3 months
Methyl. MGMT CSF Pretreatment MeDIP/PCR 66 AOA/GBM NA No NA 11.3 months
Methyl. p16 CSF Pretreatment MeDIP/PCR 66 AOA/GBM NA No NA 11.3 months
Methyl. TIMP-3 CSF Pretreatment MeDIP/PCR 66 AOA/GBM NA No NA 11.3 months
Methyl. THBS1 CSF Pretreatment MeDIP/PCR 66 AOA/GBM NA No NA 11.3 months
Wakabayashi et al 2009113 Methyl. p16 Serum Pretreatment PCR 40 Glioma NOS NA NA NA NA
Skog et al 200896 EGFRvIII Serum MVs Intraoperative RT-PCR 25 GBM NA NA NA NA
Weaver et al 2006111 Methyl. p16 Plasma Intraoperative PCR 10 AII/AIII/GBM NA NA NA NA
Methyl. MGMT Plasma Intraoperative PCR 10 AII/AIII/GBM NA NA NA NA
Methyl. p73 Plasma Intraoperative PCR 10 AII/AIII/GBM NA NA NA NA
RARbeta Plasma Intraoperative PCR 10 AII/AIII/GBM NA NA NA NA
Balana et al 2003110 Methyl. DAPK Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA
Methyl. p16 Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA
Methyl. MGMT Serum Pre/Intraoperative PCR 28 GBM Yes NA NA NA
Methyl. RASSF1A Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA
Ramirez et al 2003112 Methyl. MGMT Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA
Methyl. p16 Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA
Methyl. DAPK Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA
Methyl. RASSF1A Serum Pre/Intraoperative PCR 28 GBM NA NA NA NA

Abbreviations: AII, astrocytoma WHO grade 2; AIII, astrocytoma WHO grade III; AOA, anaplastic oligoastrocytoma; BEAMing, beads, emulsion, amplification, magnetics; CSF, cerebrospinal fluid; CTC, circulating tumor cell; DAPK, death-associated protein kinase 1; ddPCR, droplet digital PCR; EGFRvIII, endothelial growth factor variant 3; GBM, glioblastoma (or astrocytoma WHO grade IV); Glioma NOS, glioma not otherwise specified; HGG, high-grade glioma; IDH1, isocitrate dehydrogenase 1; MeDIP, methylated DNA immunoprecipitation; Methyl, methylation; MGMT, O6-alkylguanine DNA alkyltransferase; miR-, microRNA; NA, not available; OII, oligodendroglioma WHO grade II; OIII, oligodendroglioma WHO grade III; PTEN, phosphatase and tensin homolog; qRT-PCR, real-time reverse-transcription PCR; RARbeta, retinoic acid receptor beta; RASSF1A, Ras association domain-containing protein 1A; THBS1, thrombospondin1; TIMP-3, metalloproteinase inhibitor 3; TPBA, telomerase promoter-Based Assay; intraop, intraoperative.

All studies listed in Table 1 were observational studies.

Circulating Tumor-derived Exosomes

Exosomes (microvesicles or extracellular vesicles) are 30–100 nm in diameter and are released into the microenvironment of cells or into surrounding body fluids by both normal and cancer cells, where they perform a variety of functions.9193 Exosomes can be taken up by particular host cells and thus provide signaling between various cell types including cancer cells.9496 Circulating tumor-derived exosomes contain a variable spectrum of molecules representative of the parental cells including proteins, nucleic acids, lipids, metabolites, and other molecules. Cancer cell exosomes carry molecular signatures and effectors of diseases such as mutant oncoproteins, oncogenic transcripts, microRNA, and DNA sequences. Their contents can help identify the cells of origin for the exosomes, thereby offering the opportunity to identify biomarkers or therapeutic targets in body fluids.9193,97,98 Circulating exosomes in the body fluids of brain tumor patients may be used to decipher molecular features of the neoplasms or measure their responses to therapy.96,97 So far, various tumor-related molecules with altered expression patterns have been found in circulating exosomes of glioma patients including EGFRvII,96,99 EGFR,99 podoplanin (PDPN),99 phosphatase and tensin homolog (PTEN),12 miR-21,100 and mutant IDH1 mRNA (Table 1).9,99 Exosomes may carry substantial amounts of bound antibody-recognizing tumor antigens (autoantibodies), which can be used to reveal the presence of tumor antigens; exosome-based immunotherapy is under development.96,97,101,102 Although exosomes are promising targets of biomarker research, their tracing and quantification in clinical samples remain challenging.103,104 New technologies, such as ExoScreen, are being developed for eventual clinical use.103

Circulating Tumor-associated Nucleic Acids

Circulating nucleic acids (CNAs) have been identified in blood and other bodily fluids of patients with various diseases.108,118 CNAs are promising targets for development as tumor biomarkers (circulating tumor-associated nucleic acids [ctNAs]) because of the possibility to profile tumors at the genomic and transcriptomic levels. Nucleic acids appear in body fluids as a sequel of apoptotic tumor cells or tumor necrosis, but they may also be actively secreted into the circulation.105 Levels of CNAs are influ­enced by many factors: the turnover of (tumor) cell populations, cell degradation rates, filtering processes present in the blood or lymphatic circulation, clearance by liver and kidney, infection, age, sex, treatment, stress on epigenetic mechanisms, diet, lifestyle, and more.105,106 Although nucleic acids are valuable as biomarkers because they can be measured in sensitive high-throughput PCR detection assays, the identification, quantitation, and validity of ctNAs remain challenging. In order to link the presence of ctDNA, circulating tumor-associated RNA (ctRNA), or circulating tumor-associated microRNA (ctmiRNA) in body fluids of cancer patients to tumor-specific molecular events, the preanalytic conditions must be well defined and standardized.58,105

Circulating Tumor-associated DNA

ctDNA may harbor the genetic and epigenetic aberrations present in tumors and their metastases including point mutations, rearrangements, amplifications, and aneuploidy. The aberrations may be highly specific for an individual tumor and may also represent its molecular heterogeneity.107 ctDNAs have been detected in patients with tumors of breast, bladder, colon, liver, lung, ovaries, pancreas, and prostate as well as non-Hodgkin's lymphoma and melanoma. ctDNAs have also been detected in glioma patients, and the aberrations found included IDH1 mutation,108 loss of heterozygosity for 1p, 10q, 19q12; EGFRvIII mutation;109 as well as abnormal methylation of the promoters of MGMT1113,110112, p16,110113 DAPK,110,112 RASSF1A,13,110,112 p73,111 RARbeta,111 PTEN,12 p15INK4B,13 and p14ARF.13 At this point, the clinical utility has not been validated for any of the candidate ctDNAs as biomarkers for glioma patients. Prospective settings are needed for clinically applicable tests.

Circulating tumor-associated RNA and microRNA

The characterization of ctRNAs has not been explored to the extent of ctDNA, and the tumor specificity of these CNAs has been challenged more vigorously. One reason is that cell-free RNA is prone to degradation by the ubiquitous presence of RNA-degrading enzymes, which are generally elevated in the serum of cancer patients.114,115 Extracellular RNA is usually present in the exosomes.116 Aberrant RNA expression has been associated with stage, progression, and spread of various cancer types.117 In patients with gliomas, exosomes and platelets have been used as sources for detecting tumor-associated RNA profiles, among which are mutant EGFRvIII and mutant IDH1.9,96,116,118 As with the ctDNAs, the ctRNAs have also not been validated as biomarkers for introduction into clinical practice.

microRNAs (miRNAs) are noncoding, single-stranded RNAs of ∼22 nucleotides that constitute a novel class of gene regulators and function as tumor suppressors and oncogenes.119 Because miRNAs, unlike RNA, are relatively stable and are present in blood and other bodily fluids, they are potential tumor biomarkers and may be more sensitive and specific for detecting tumors than currently available methods for early diagnosis of cancer.120 The peripheral blood contains large amounts of stable miRNAs derived from various tissues, and alterations in these miRNA have been reported for many tumors including gliomas.119129 Deviant miRNA expression patterns in the blood of glioma patients include miR-10b,130 miR-15b,106,131,132 miR-17–5p,106 miR-20a,106 miR-21,96,130,132135 miR-23a,106,131 miR-31,106 miR-106,106 miR-128,106,135,136 miR-133a,131 miR-146b,106 miR-148a,106 miR-150,106,131 miR-193a,106 miR-197,131miR-200b,106 miR-221,106 miR-222,106 miR-342-3p,135,136 miR-497,131 and miR-548b-5p.131 Some significant technological pitfalls and limitations need to be addressed before the miRNAs can be introduced as clinically applicable glioma biomarkers.124,137

Circulating Tumor-associated Protein Biomarkers

Efforts have been made over the last decades to identify candidate protein biomarkers for gliomas that could be measured in body fluids (eg, urine, serum/plasma, or CSF) for making a diagnosis, detecting recurrence, or monitoring tumor activity following therapy (Table 2).32,33,3639,4143,45,4751,138155 Recent advances in proteomics have led to an explosion of activity in the field of biomarker research, particularly that related to cancers. The identification of biomarkers in body fluids such as serum is difficult due to the large dilution factor and the abundance of other constitutive serum proteins. Sample enrichment is necessary to enhance the sensitivity, and extensive validation of the methodology is necessary to ensure the specificity of candidate biomarkers.156 Several reports on the analysis of the glioma proteome, in which tumor tissue, serum, plasma, CSF or cyst fluids have been implicated.34,36,157161 Attempts have also been made to identify biomarkers by using xenografts in animal models.162

Table 2.

Potential glioma protein biomarkers in cerebrospinal fluid, serum and plasma

Reference Biomarker Study Type Source Time of Take Methodology n Tumor Type Treatment Response Survival Radiology Median Follow-up
Yamaguchi et al 201330 OPN Obser CSF Pretreatment ELISA 63 GBM/metastases NA NA NA NA
Verschuere et al 2013258 Galectin-1 Obser serum Intraop./postoperative ELISA 125 HGG/recurrent HGG NA NA NA NA
Chinnaiyan et al 2012150 IGFBP-5 Inter Phase I plasma Pre/posttreatment ELISA 10 Recurrent GBM Yes Yes NA 5.7 months
PDGF Inter Phase I plasma Pre/posttreatment ELISA 10 Recurrent GBM Yes Yes NA 5.7 months
Li et al 2012149 IGFBP-2 Obser serum Preoperative ELISA 145 Glioma NOS NA NA NA NA
Bemardi et al 2012235 YKL-40 Obser Serum Postoperative ELISA 60 GBM Yes Yes NA 12 months
Iwamoto et al 2011218 MMP-9 Obser Serum Pre/posttreatment ELISA 343 AII/AIII/GBM NA No No 29 to 52 months
Li et al 2011149 IGFBP-2 Obser CSF Preoperative ELISA 94 AGG/carcinomas NA Yes NA 7–24 months
Mittelbronn et al 201132 MIF Obser CSF Pretreatment ELISA 14 HGG NA NA NA NA
Schuhmann et al 201033 OPN Basic CSF Preoperative MALDI-TOF-MS 11 GBM No No NA NA
AACT Basic CSF Preoperative MALDI-TOF-MS 11 GBM No No NA NA
TTHY Basic CSF Preoperative MALDI-TOF-MS 11 GBM No No NA NA
ALB Basic CSF Preoperative MALDI-TOF-MS 11 GBM No No NA NA
Batchelor et al 2010194 VEGF Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes No No 227 days
PlGF Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes Yes Yes 227 days
VEGFR2 Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes No No 227 days
FGF-ß Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes Yes Yes 227 days
MMP2 Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes Yes Yes 227 days
MMP10 Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes No No 227 days
SDF-1a Inter Phase II Serum Pre/post-treatment ELISA 31 Recurrent GBM Yes No No 227 days
Tie2 Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes No No 227 days
Agn2 Inter Phase II Serum Pre/posttreatment ELISA 31 Recurrent GBM Yes No No 227 days
Sreekanthreddy et al 2010207 OPN Obser Serum Preoperative ELISA+WB 30 GBM NA Yes NA 34 months
Ohnishi et al 2009259 Gelsolin Basic CSF Pretreatment MALDI-TOF-MS 2 AII/GBM NA NA NA NA
Lin et al 2009195 IGFBP-2 Obser Plasma Pre/posttreatment ELISA 196 AGG NA Yes NA 1 year
Ilhan et al 2009182 Ang2 Obser Plasma Pretreatment ELISA 78 AGG/metastases No Yes NA 8 months
VEGF Obser Plasma Pretreatment ELISA 78 AGG/metastases No Yes NA 8 months
PDGF Obser Plasma Pretreatment ELISA 78 AGG/metastases No Yes NA 8 months
Petrik et al 2008260 AHSG Obser Serum Preoperative SELDI-TOF, ELISA 214 AII/AIII/GBM No Yes NA 2 years
Reddy et al 2008261 PBEF1 Obser Serum Preoperative ELISA 95 AIII/GBM NA Yes NA 8 months
Iwadate et al 2008262 PAI-1 Obser Serum Pre/posttreatment ELISA 57 AGG NA Yes NA NA
Jung et al 2007143 GFAP Obser Serum Pretreatment ELISA 104 AGG/metastases No NA NA NA
Brommeland et al 2007226 GFAP Obser Serum Preoperative ELISA 31 HGG/metastases No NA Yes NA
Todaro et al 2007263 NCAM Obser Serum Pretreatment WB 61 AGG/metastases Yes No NA 1–3 months
Zheng et al 2007155 eNOS Obser Plasma Pretreatment ELISA 115 AGG/metastases NA NA NA NA
Quaranta et al 2007264 EGFR Obser Serum Pre/postoperative ELISA 65 HGG NA Yes NA 13 months
Khwaja et al 200635 Attractin Obser CSF Pretreatment 2-DE + cICAT 47 Glioma NOS/metastases NA NA NA NA
Hormigo et al 2006219 YKL-40 Obser Serum Pre/postoperative ELISA 143 HGG NA Yes Yes 4.6 to 22 months
MMP9 Obser Serum Pre/postoperative ELISA 143 HGG NA No No 4.6 to 22 months
Ilzecka et al 2006265 APRIL Obser Serum Preoperative ELISA 25 GBM NA NA No NA
Zheng et al 2005198 L-CaD Obser Serum Pretreatment ELISA 57 AGG No NA NA NA
Fukuda et al 2005266 Cathepsin D Obser Serum Pre/post-operative ELISA 20 AGG NA NA NA NA
Peles et al 200438 VEGF Obser CSF Pre/Intraoperative ELISA 26 AGG NA Yes NA 652 days
VEGF Obser Serum Pre/Intraoperative ELISA 26 AGG NA No NA 652 days
FGF-ß Obser CSF Pre/Intraoperative ELISA 26 AGG NA Yes NA 652 days
FGF-ß Obser Serum Pre/Intraoperative ELISA 26 AGG NA No NA 652 days
Sampath et al 200439 VEGF Obser CSF Pre/Intraoperative ELISA 27 AII/AIII/metastases NA NA NA NA
Recoverin Obser serum Pre/Intraoperative ELISA 24 AGG NA NA NA NA
Zheng et al 200340 L-CaD Basic CSF Intraoperative 2D + MALDI 10 AGG No NA NA NA
Batabyal et al 200337 CEA Obser CSF Pretreatment RIA 22 AGG/metastases No NA NA NA
Ribom et al 200341 PDGF Obser CSF Pre/postoperative Radioreceptor assay 7 LGG NA NA NA NA
VEGF Obser CSF Pre/postoperative ELISA 7 LGG NA NA NA NA
VEGF Obser Serum Pre/postoperative ELISA 7 LGG NA NA NA NA
FGF-ß Obser CSF Pre/postoperative ELISA 7 LGG NA NA NA NA
FGF-ß Obser serum Pre/postoperative ELISA 7 LGG NA NA NA NA
Tanwar et al 2002256 YKL-40 Obser Serum NA ELISA 65 AGG NA NA NA NA
Fine et al 2000138 FGF-ß Inter Phase II Serum Pre/posttreatment ELISA 39 HGG Yes Yes Yes 80 weeks
VEGF Inter Phase II Serum Pre/post-treatment ELISA 39 HGG Yes No No 80 weeks
Streffer et al 199842 CD95 Obser CSF Pretreatment ELISA 20 GBM NA NA NA NA
Rudenko et al 1996230 CEA Obser CSF Preoperative SFI 83 Brain tumors NOS NA NA NA NA
CEA Obser Serum Preoperative SFI 83 Brain tumors NOS NA NA NA NA
AFP Obser CSF Preoperative SFI 83 Brain tumors NOS NA NA NA NA
AFP Obser Serum Preoperative SFI 83 Brain tumors NOS NA NA NA NA
HCG Obser CSF Preoperative SFI 83 Brain tumors NOS NA NA NA NA
HCG Obser Serum Preoperative SFI 83 Brain tumors NOS NA NA NA NA
CEA Obser Serum Pretreatment SFI 101 Brain tumors NOS NA NA NA NA
Rombos et al 1988141 CEA Obser CSF Pre/postoperative SFI 41 AGG/metastases No NA NA NA
CEA Obser Serum Pre/postoperative SFI 41 AGG/metastases No NA NA NA
AFP Obser CSF Pre/postoperative SFI 41 AGG/metastases No NA NA NA
AFP Obser Serum Pre/postoperative SFI 41 AGG/metastases No NA NA NA
Suzuki et al 198050 CEA Obser CSF Pre/posttreatment RIA 253 AGG/metastases Yes NA NA NA
CEA Obser Plasma Pre/posttreatment RIA 253 AGG/metastases Yes NA NA NA
Miyake et al 197951 CEA Obser CSF Pre/posttreatment RIA 97 AGG/metastases Yes NA NA NA

Abbreviations: AGG, all grade of gliomas; AII, astrocytoma WHO grade 2; AIII, astrocytoma WHO grade III; AACT, alpha-1-antichymotrypsin; ABL, N-terminal residue of albumin; AHSG, 2-Heremans-Schmid glycoprotein; Ang2, angiopoietin2; APRIL, a proliferation-inducing ligand; CSF, cerebrospinal fluid; EGFR, epidermal growth factor receptor; eNOS, endothelial nitric oxide synthase; FGF-β, basic fibroblast growth factor; GBM, glioblastoma; GFAP, glial fibrillary acidic protein; HGG, high-grade glioma; IGFBP-2, insulin-like growth factor-binding protein 2; IGFBP-5, insulin-like growth factor-binding protein 5; intraop, intraoperative; L-CaD, low molecular; LGG, low-grade glioma; MIF, macrophage migration inhibitory factor; MMP2/9/10, matrix metalloproteinase-2/9/10; NCAM, neural cell adhesion molecule; NOS, not otherwise specified; Obser, observational studies; OPN, osteopontin; PAI-1, plasminogen activator inhibitor 1; PBEF1, pre-B-cell colony enhancing factor 1; PDGF, platelet-derived growth factor; PlGF, placental growth factor; SDF-1α, stromal cell-derived factor 1; Tie2, angiopoietin receptors; TTHY, transthyretin; VEGF, vascular endothelial growth factor; VEGFR2, vascular endothelial growth factor receptor 2; Wb, Western blot; YKL-40, (tyrosine (Y), lysine (K) and leucine (L) and the apparent molecular weight).

Growth Factors and Angiogenesis-related Biomarkers

Vascular Endothelial Growth Factor

Gliomas are highly vascularized tumors, and the process of angiogenesis is progressive throughout tumor development. Obviously, the newly formed vessels are attractive targets for antiangiogenic therapy. Vascular endothelial growth factor (VEGF) is a key molecule for triggering the process of angiogenesis in pathological conditions including neoplasms.163 Tumor hypoxia due to increased cell density triggers the angiogenic switch by upregulating VEGF.164 Given the importance of VEGF in tumor angiogenesis, several drugs to suppress VEGF signaling have been developed.165 Bevacizumab is the most well-characterized antiangiogenic drug currently being used for the treatment of human GBM. Bevacizumab is a humanized monoclonal antibody that binds to circulating VEGF and prevents its interaction with the VEGF receptor suppressing VEGF signaling.166172 Antiangiogenic strategies are targeted to endothelial cells, although the main sources of VEGF are the glial tumor cells.165,173 Patients usually become resistant to anti-VEGF therapy after an initial response due to various compensation mechanisms.165,169,174176 VEGF has been considered to be a potential protein biomarker in CSF and serum/plasma of glioma patients, and its elevated levels have appeared to correlate with the microvascular density of the tumors.38,39,177182 Because VEGF levels of serum are also increased in other systemic cancers, including breast cancers,183185 lung cancer,186,187 and colon cancer,188190 they are not specific to glial tumors. In several clinical studies, it was demonstrated that the serum VEGF levels of cancer patients, including gliomas, remained significantly high following surgery, radiotherapy, or chemotherapy.179,191 Moreover, particular inhibitors of the VEGF receptor tyrosine kinases induce increased levels of serum VEGF.192 Taken together, the value of VEGF as a biomarker has not been established in glioma.

Other Growth Factors and Angiogenesis-associated Molecules

Aside from VEGF, various growth factors and other angiogenesis-associated molecules have been used to monitor the effects of antiangiogenic therapy in gliomas.38,138,155,168,193,194 Growth factors are potential targets for therapeutic strategies because they are essential for tumor progression. Changes in plasma placental growth factor, basic fibroblast growth factor (FGF-β), soluble VEGF receptor 1, soluble VEGF receptor 2, stromal cell-derived factor-1alpha (SDF-1alpha), and soluble Tek/Tie2 receptor were all used to monitor the effects of cediranib (a pan-VEGF receptor tyrosine kinase inhibitor) in several clinical studies.168,193,194 All of these molecules reportedly correlated with radiological response and overall survival.194 FGF-β levels related to tumor progression and overall survival were also evaluated apart from the cediranib trial.38,138 The factors or molecules evaluated in more than one study include platelet-derived growth factor (PDGF),41,150,182 insulin-like growth factor binding protein 2 (IGFBP-2),149,195 and angiopoietin2 (Ang2)182,194 (Table 1). Endothelial nitric oxide synthase (eNOS), a specific isoform of the nitric oxide-producing enzyme of endothelial cells (ECs), is a well-characterized marker of ECs.196 This molecule is activated in the process of angiogenesis and vasculogenesis and plays an intimate role in VEGF signaling.197 The blood level of eNOS largely reflects the activity of cells with endothelial lineage.196,197 The concentration of plasma eNOS is significantly greater in glioma patients as compared with control groups.155 The expression of low molecular isoform of caldesmon (l-CaD), a cytoskeleton-associated protein, was also increased in CSF40 and serum198 in glioma patients. The expression of l-CaD in blood vessels was further confirmed in tissue sections of glioma patients.199202 A zebrafish l-CaD knockdown model further confirmed that this molecule plays a crucial role in vasculogenesis and angiogenesis in vivo.203 So far, the performance of l-CaD and eNOS as biomarkers for glioma has not been tested in additional studies.

Matricellular Proteins

The group of matricellular proteins consists of structurally diverse glycoproteins that are secreted by tumor cells and neighboring stromal cells.204206 These proteins are secreted into the extracellular environment, and they interact with cell-surface receptors, proteases, hormones, and structural matrix proteins such as collagens.205 Matricellular proteins are also involved in various aspects of tumor biology such as EMT, angiogenesis, cell proliferation and survival, motility, and ECM degradation.204206 Glial tumor cells need to break down the environmental substances in order to infiltrate diffusely into the surrounding brain tissue. Among the many proteins that serve in this context are thrombosponin-1 and-2 (TSP1, TSP2), tenascin-C (TNC), secreted protein acidic and rich in cysteine (SPARC), osteopontin (OPN), angiopoietin-like protein 4 (ANGPTL4), CCN family members cysteine-rich angiogenic inducer 61 (Cyr61/CCN1) and CCN6, periostin, and more.204 Some of these proteins have been scrutinized for their value as glioma biomarkers in CSF and serum (eg, OPN30,33,207 and tenascin).43 CSF levels of tenascin were reportedly higher in anaplastic gliomas as compared with astrocytomas of lower malignancy.43 Increased expression of OPN has been associated with the presence of a variety of cancers including breast cancer, ovarian cancer, melanoma, and glioblastoma.208 The presence of metastases was also found to be associated with high OPN levels.209,210 CSF and serum levels of OPN appeared to be higher in patients with gliomas as compared with patients with other primary brain tumors or systemic cancers, and the levels were associated with worse outcomes.30,33,207 However, no correlation between the radiographic properties of the tumor and the OPN level was found. Interestingly, significant differences in OPN levels between patients with gliomas of WHO grades II, III, and IV were found, and the survival times of patients with high serum OPN levels (>20 ng/mL) appeared to be significantly shorter than those of patients with low OPN levels. Postoperative levels of OPN were, however, not measured in these studies.30,33,207 So far, the value of OPN for monitoring treatment response is unclear. Since OPN levels were reportedly higher in CSF of patients with atypical teratoid/rhabdoid tumors211 and other tumors,208 OPN is not specific for glioma and cannot be used as a diagnostic biomarker.

Matrix Metalloproteinase

Matrix metalloproteinases (MMPs) represent a family of degrading enzymes involved in the breakdown of extracellular matrices necessary for invasion of tumor cells.212 The zinc- and calcium-dependent MMP family also plays a role in various physiological processes such as embryonic development, angiogenesis, wound healing, and more.212,213 MMPs comprise a relatively large and ever-growing family, and more than 20 enzymes are now known.212 MMP-2 (gelatinase-A) and MMP-9 (gelatinase-B) are the most abundant MMPs in malignant gliomas.214216 In glial tumors, MMP-9 in particular enables tumor cells to migrate or infiltrate, and its level is upregulated by the expression of astrocyte elevated gene-1 (AEG-1).217 MMP-9 has been measured in serum218,219 and CSF220,221 in glioma patients. Levels of MMP-9 have been measured in the CSF of patients treated for glioma recurrence, and elevated concentrations were interpreted to be indicative of treatment failure.220 The presence of activated MMP-9 also correlated with positive CSF cytology.221 There are, however, conflicting results for the value of MMP-9 levels in serum.218,219 In a longitudinal study with a larger patient population, no statistically significant association was observed between levels of serum MMP-9 and radiographic disease status, and changes in serum MMP-9 did not appear to be associated with survival in a cohort of patients with anaplastic glioma.218 In contrast, serum MMP-9 levels were associated with disease status and were inversely correlated with prognosis in 77 GBM and 66 anaplastic glioma patients.219 A caveat of these studies is the fact that leukocytes secrete MMP-9, and increased numbers of leukocytes in CSF or serum can cause increased levels of MMP-9.222224 Serum or CSF levels of MMP-9 in glioma patients may therefore be influenced by concomitant inflammation. Because MMPs are crucial for angiogenesis, tumor invasion, tumor growth, and metastatic potential, MMPs are promising targets for potential therapies.225

Proteins Associated With Cell Lineage

Glial Fibrillary Acidic Protein

Glial fibrillary acidic protein (GFAP) is an intermediate filament-associated protein, and its immunohistochemistry is used for revealing astrocytic lineage of glial cells and glial tumor cells. Serum levels of GFAP have been analyzed in several clinical studies143,226,227 and were significantly elevated in high-grade gliomas, as compared with those of nonglial tumors, with 100% specificity for the diagnosis of gliomas.143,226,227 Jung et al prospectively examined 50 patients with GBM, 18 with anaplastic gliomas, 13 with low-grade gliomas, 17 with a single cerebral metastasis, and 50 healthy controls.143 Serum GFAP levels were measured by ELISA and were detectable in 40 of the 50 GBM patients (median, 0.18 µg/L; range, 0–5.6 µg/L). Only 2 patients with gliomas of low malignancy grade had detectable serum GFAP levels. Serum GFAP levels in patients with metastases and healthy people were below the detection limit (≤0.012 µg/L). The GFAP serum levels correlated with both tumor volumes and estimated volumes of tumor necrosis in the GBM patients. Brommeland et al found GFAP serum levels with a broad range from 30–1210 ng/L (mean, 239 ng/L) and demonstrated a significant association between preoperative serum GFAP levels and tumor volume in 31 high-grade glioma patients by using ELISA.226 An ELISA has been recently developed for an autoantibody to GFAP, to be used for detection of glioma.228 The GFAP serum level may well become a useful protein biomarker. At this point, GFAP should be validated in appropriate clinical studies.

Embryonic Antigens

Carcinoembryonal antigen (CEA), human chorionic gonadotropin (hCG) and alpha-fetoprotein (AFP) are useful markers for the differential diagnosis between primary brain tumors and metastases or germ cell tumors (GCTs). The cell adhesion molecule CEA is an embryonic antigen and is produced in gastrointestinal tissues during fetal development.229 Levels of CEA were monitored in the serum, CSF, plasma, and tumor cyst fluid48,50,51,141,230 of patients with primary brain tumors and cerebral metastases. CEA levels in patients with cerebral metastases and leptomeningeal dissemination were consistently higher than those in patients with primary brain tumors.37,45,48,51,141 In a study with postoperative follow-up, it was found that patients with metastatic brain tumors and leptomeningeal tumor spread showed high levels of CEA in CSF preoperatively (which normalized following surgery).45 These data support the use of CEA levels in CSF for the differential diagnosis of primary and metastatic brain tumors.37 The detection of hCG in serum or CSF supports the diagnosis of intracranial GCTs and proves the presence of trophoblastic cells.231 AFP is an oncofetal glycoprotein that plays an important role during embryo- and fetogenesis. Elevated serum AFP concentrations have been associated with hepatocellular carcinoma and pediatric tumors,231 and measurement of hCG and AFP in CSF or serum is considered to be of significance in the differential diagnosis of GCTs and other tumors including glioma.147,148,231

Miscellaneous Proteins and Circulating Oncometabolites

2-hydroxyglutamate

Cancer-associated IDH mutations produce the metabolite 2-hydroxyglutarate (2HG). Circulating levels of 2HG are significantly elevated in patients with cholangiocarcinoma232 and acute myeloid leukemia.233 In a recent study, it was reported that the concentration of the metabolite 2HG in serum from glioma patients did not correlate with the IDH1 mutational status or the size of the tumor.234 More clinical studies are required to evaluate the clinical utility of 2HG.

YKL-40

YKL-40 (tyrosine (Y), lysine (K) and leucine (L) and the apparent molecular weight) is also known as chitinase-3-like-1 or human cartilage glycoprotein-39.235,236 There are indications that YKL-40 may promote degradation of the ECM and play a role in cell migration,237 connective tissue modeling,238240 and inflammatory responses.241,242 Serum levels of YKL-40 are elevated in patients with lymphoma,243 lung cancer,244 leukemia,245 melanoma,246,247 colon cancer,248,249 ovarian cancer,250252 breast carcinoma,253,254 and prostate cancer,255 and raised levels of YKL-40 correlate with shorter survival times. YKL-40 is highly expressed in tissue microarray studies of gliomas.256 In multivariate analysis, the tissue expression of YKL-40 was identified as an independent predictor of survival after adjusting for patient age, performance status, and extent of resection.257 The expression of YKL-40 appeared to be associated with loss of chromosome arm 10q.236 YKL-40 is secreted into the bloodstream by tumor cells and tumor-associated macrophages and can be detected by ELISA.256 The value of YKL-40 as a serum marker was evaluated during a follow-up period of 27 months after surgery for high-grade glioma.219 Levels of YKL-40 were significantly correlated with radiographic evidence of disease and survival times in GBM (n = 76) and anaplastic glioma (n = 66).219 In a prospective study in which 1740 MRI matched serum samples of 343 anaplastic glioma patients were implicated, the YKL-40 levels appeared to be significantly lower in patients with no radiographic tumor progression as compared with patients with progressive disease.257 Increases in YKL-40 levels were also associated with worse survival.257 In various other clinical studies, serum YKL-40 levels of glioma patients were also elevated and correlated with radiographic evidence of disease and worse overall survival.219,235,256,257 Since serum levels of YKL-40 are also correlated with poor outcome in various cancers,243245,249255 additional validation studies need to be done, focusing on the specificity of YKL-40 and testing of its value as a glioma biomarker in prospective, controlled settings.

Other proteins

Various other proteins, which are not discussed here, are listed in Table 1 and include galectin-1,258 nerve growth factor (NGF),31 macrophage migration inhibitory factor (MIF),32 alpha-1-antichymotrypsin (AACT),33 transthyretin (TTHY),33 gelsolin,259 2-Heremans-Schmid glycoprotein (AHSG),260 Pre-B-cell colony enhancing factor 1 (PBEF1),261 plasminogen activator inhibitor 1 (PAI-1),262 neural cell adhesion molecule (NCAM),263 EGFR,264 attractin,35 a proliferation-inducing ligand (APRIL),265 Cathepsin D,266 recoverin,39 CD95,42 G-22,267 and somatomedins.47

Concluding Remarks

Current strategies in the therapy for patients suffering from primary brain tumors necessitate the development of practical and standardized assays for monitoring disease activity and therapy effects. Intracranial tumors are not accessible for frequent sampling, and therefore body fluids such as blood and CSF are preferable sources for biomarkers. A large number of candidate biomarkers have been discovered, but neither circulating tumor cells, nor their exosomes, DNA, RNA, and particular proteins have passed the requirements of the Tumor Marker Utility Grading System Levels of Evidence/NCCN for clinical application or for serving as monitors in trials. The road from the discovery of new candidate biomarkers to their clinical validation is long. Many issues need to be addressed including biological relevance, sensitivity, specificity, and reproducibility of the measurements. Technical standardization is crucial to achieve clinical utility for candidate biomarkers. Collaborating consortia are needed for standardization and validation of sample collection and isolation, and large prospective multicenter studies are needed to reach the level of evidence required for introducing new biomarkers into clinical practice.

Funding

None declared.

Acknowledgments

None declared.

Conflict of interest statement. None declared.

References

  • 1.Parkin DM, Bray F, Ferlay J, et al. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55(2):74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 2.Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109. doi: 10.1007/s00401-007-0243-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ohgaki H, Kleihues P. Genetic pathways to primary and secondary glioblastoma. Am J Pathol. 2007;170(5):1445–1453. doi: 10.2353/ajpath.2007.070011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol. 2005;64(6):479–489. doi: 10.1093/jnen/64.6.479. [DOI] [PubMed] [Google Scholar]
  • 5.Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765–773. doi: 10.1056/NEJMoa0808710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.van den Bent MJ, Dubbink HJ, Marie Y, et al. IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: a report of the European Organization for Research and Treatment of Cancer Brain Tumor Group. Clin Cancer Res. 2010;16(5):1597–1604. doi: 10.1158/1078-0432.CCR-09-2902. [DOI] [PubMed] [Google Scholar]
  • 7.van den Bent MJ, Dubbink HJ, Sanson M, et al. MGMT promoter methylation is prognostic but not predictive for outcome to adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors: a report from EORTC Brain Tumor Group Study 26951. J Clin Oncol. 2009;27(35):5881–5886. doi: 10.1200/JCO.2009.24.1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.van den Bent MJ, Taphoorn MJ, Brandes AA, et al. Phase II study of first-line chemotherapy with temozolomide in recurrent oligodendroglial tumors: the European Organization for Research and Treatment of Cancer Brain Tumor Group Study 26971. J Clin Oncol. 2003;21(13):2525–2528. doi: 10.1200/JCO.2003.12.015. [DOI] [PubMed] [Google Scholar]
  • 9.Chen WW, Balaj L, Liau LM, et al. BEAMing and Droplet Digital PCR Analysis of Mutant IDH1 mRNA in Glioma Patient Serum and Cerebrospinal Fluid Extracellular Vesicles. Mol Ther Nucleic Acids. 2013;2:e109. doi: 10.1038/mtna.2013.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Liu BL, Cheng JX, Zhang W, et al. Quantitative detection of multiple gene promoter hypermethylation in tumor tissue, serum, and cerebrospinal fluid predicts prognosis of malignant gliomas. Neuro Oncol. 2010;12(6):540–548. doi: 10.1093/neuonc/nop064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Balana C, Carrato C, Ramirez JL, et al. Tumour and serum MGMT promoter methylation and protein expression in glioblastoma patients. Clin Transl Oncol. 2011;13(9):677–685. doi: 10.1007/s12094-011-0714-x. [DOI] [PubMed] [Google Scholar]
  • 12.Lavon I, Refael M, Zelikovitch B, et al. Serum DNA can define tumor-specific genetic and epigenetic markers in gliomas of various grades. Neuro Oncol. 2010;12(2):173–180. doi: 10.1093/neuonc/nop041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Majchrzak-Celinska A, Paluszczak J, Kleszcz R, et al. Detection of MGMT, RASSF1A, p15INK4B, and p14ARF promoter methylation in circulating tumor-derived DNA of central nervous system cancer patients. J Appl Genet. 2013;54(3):335–344. doi: 10.1007/s13353-013-0149-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28(11):1963–1972. doi: 10.1200/JCO.2009.26.3541. [DOI] [PubMed] [Google Scholar]
  • 15.Macdonald DR, Cascino TL, Schold SC, Jr., et al. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8(7):1277–1280. doi: 10.1200/JCO.1990.8.7.1277. [DOI] [PubMed] [Google Scholar]
  • 16.Chinot OL, Macdonald DR, Abrey LE, et al. Response assessment criteria for glioblastoma: practical adaptation and implementation in clinical trials of antiangiogenic therapy. Curr Neurol Neurosci Rep. 2013;13(5):347. doi: 10.1007/s11910-013-0347-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Brandsma D, Stalpers L, Taal W, et al. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453–461. doi: 10.1016/S1470-2045(08)70125-6. [DOI] [PubMed] [Google Scholar]
  • 18.Brandsma D, van den Bent MJ. Pseudoprogression and pseudoresponse in the treatment of gliomas. Curr Opin Neurol. 2009;22(6):633–638. doi: 10.1097/WCO.0b013e328332363e. [DOI] [PubMed] [Google Scholar]
  • 19.de Wit MC, de Bruin HG, Eijkenboom W, et al. Immediate post-radiotherapy changes in malignant glioma can mimic tumor progression. Neurology. 2004;63(3):535–537. doi: 10.1212/01.wnl.0000133398.11870.9a. [DOI] [PubMed] [Google Scholar]
  • 20.Taal W, Brandsma D, de Bruin HG, et al. Incidence of early pseudo-progression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomide. Cancer. 2008;113(2):405–410. doi: 10.1002/cncr.23562. [DOI] [PubMed] [Google Scholar]
  • 21.Kruser TJ, Mehta MP, Robins HI. Pseudoprogression after glioma therapy: a comprehensive review. Expert Rev Neurother. 2013;13(4):389–403. doi: 10.1586/ern.13.7. [DOI] [PubMed] [Google Scholar]
  • 22.Engh JA. Differential scanning calorimetry applied to cerebrospinal fluid analysis in glioblastoma. Neurosurgery. 2011;69(4):N22–N23. doi: 10.1227/01.neu.0000405600.31826.70. [DOI] [PubMed] [Google Scholar]
  • 23.Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS. 2010;5(6):463–466. doi: 10.1097/COH.0b013e32833ed177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69(3):89–95. doi: 10.1067/mcp.2001.113989. [DOI] [PubMed] [Google Scholar]
  • 25.Hayes DF, Bast RC, Desch CE, et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst. 1996;88(20):1456–1466. doi: 10.1093/jnci/88.20.1456. [DOI] [PubMed] [Google Scholar]
  • 26.Febbo PG, Ladanyi M, Aldape KD, et al. NCCN Task Force report: Evaluating the clinical utility of tumor markers in oncology. J Natl Compr Canc Netw. 2011;9(Suppl 5):S1–32. doi: 10.6004/jnccn.2011.0137. quiz S33. [DOI] [PubMed] [Google Scholar]
  • 27.Berghoff AS, Stefanits H, Heinzl H, et al. Clinical Neuropathology Practice News 4–2012: levels of evidence for brain tumor biomarkers. Clin Neuropathol. 2012;31(4):206–209. doi: 10.5414/NP300511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Schutzer SE, Liu T, Natelson BH, et al. Establishing the proteome of normal human cerebrospinal fluid. PLoS One. 2010;5(6):e10980. doi: 10.1371/journal.pone.0010980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Stoop MP, Coulier L, Rosenling T, et al. Quantitative proteomics and metabolomics analysis of normal human cerebrospinal fluid samples. Mol Cell Proteomics. 2010;9(9):2063–2075. doi: 10.1074/mcp.M110.000877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yamaguchi Y, Shao Z, Sharif S, et al. Thrombin-cleaved fragments of osteopontin are overexpressed in malignant glial tumors and provide a molecular niche with survival advantage. J Biol Chem. 2013;288(5):3097–3111. doi: 10.1074/jbc.M112.362954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Li QY, Yang Y, Zhang Y, et al. Nerve growth factor expression in astrocytoma and cerebrospinal fluid: a new biomarker for prognosis of astrocytoma. Chin Med J (Engl) 2011;124(14):2222–2227. [PubMed] [Google Scholar]
  • 32.Mittelbronn M, Platten M, Zeiner P, et al. Macrophage migration inhibitory factor (MIF) expression in human malignant gliomas contributes to immune escape and tumour progression. Acta Neuropathol. 2011;122(3):353–365. doi: 10.1007/s00401-011-0858-3. [DOI] [PubMed] [Google Scholar]
  • 33.Schuhmann MU, Zucht HD, Nassimi R, et al. Peptide screening of cerebrospinal fluid in patients with glioblastoma multiforme. Eur J Surg Oncol. 2010;36(2):201–207. doi: 10.1016/j.ejso.2009.07.010. [DOI] [PubMed] [Google Scholar]
  • 34.Khwaja FW, Nolen JD, Mendrinos SE, et al. Proteomic analysis of cerebrospinal fluid discriminates malignant and nonmalignant disease of the central nervous system and identifies specific protein markers. Proteomics. 2006;6(23):6277–6287. doi: 10.1002/pmic.200600135. [DOI] [PubMed] [Google Scholar]
  • 35.Khwaja FW, Duke-Cohan JS, Brat DJ, et al. Attractin is elevated in the cerebrospinal fluid of patients with malignant astrocytoma and mediates glioma cell migration. Clin Cancer Res. 2006;12(21):6331–6336. doi: 10.1158/1078-0432.CCR-06-1296. [DOI] [PubMed] [Google Scholar]
  • 36.Khwaja FW, Reed MS, Olson JJ, et al. Proteomic identification of biomarkers in the cerebrospinal fluid (CSF) of astrocytoma patients. J Proteome Res. 2007;6(2):559–570. doi: 10.1021/pr060240z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Batabyal SK, Ghosh B, Sengupta S, et al. Cerebrospinal fluid and serum carcinoembryonic antigen in brain tumors. Neoplasma. 2003;50(5):377–379. [PubMed] [Google Scholar]
  • 38.Peles E, Lidar Z, Simon AJ, et al. Angiogenic factors in the cerebrospinal fluid of patients with astrocytic brain tumors. Neurosurgery. 2004;55(3):562–567. doi: 10.1227/01.neu.0000134383.27713.9a. discussion 567–568. [DOI] [PubMed] [Google Scholar]
  • 39.Sampath P, Weaver CE, Sungarian A, et al. Cerebrospinal fluid (vascular endothelial growth factor) and serologic (recoverin) tumor markers for malignant glioma. Cancer Control. 2004;11(3):174–180. doi: 10.1177/107327480401100305. [DOI] [PubMed] [Google Scholar]
  • 40.Zheng PP, Luider TM, Pieters R, et al. Identification of tumor-related proteins by proteomic analysis of cerebrospinal fluid from patients with primary brain tumors. J Neuropathol Exp Neurol. 2003;62(8):855–862. doi: 10.1093/jnen/62.8.855. [DOI] [PubMed] [Google Scholar]
  • 41.Ribom D, Larsson A, Pietras K, et al. Growth factor analysis of low-grade glioma CSF: PDGF and VEGF are not detectable. Neurol Sci. 2003;24(2):70–73. doi: 10.1007/s100720300075. [DOI] [PubMed] [Google Scholar]
  • 42.Streffer JR, Schuster M, Zipp F, et al. Soluble CD95 (Fas/APO-1) in malignant glioma: (no) implications for CD95-based immunotherapy? J Neurooncol. 1998;40(3):233–235. doi: 10.1023/a:1006173019048. [DOI] [PubMed] [Google Scholar]
  • 43.Yoshida J, Wakabayashi T, Okamoto S, et al. Tenascin in cerebrospinal fluid is a useful biomarker for the diagnosis of brain tumour. J Neurol Neurosurg Psychiatry. 1994;57(10):1212–1215. doi: 10.1136/jnnp.57.10.1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Nakagawa H, Fujita T, Tsuruzono K, et al. [Myelin basic protein in the cerebrospinal fluid of patients with neurological disease: especially with malignant brain tumors] No Shinkei Geka. 1994;22(2):111–118. [PubMed] [Google Scholar]
  • 45.Nakagawa H, Kubo S, Murasawa A, et al. [Measurements of CSF biochemical tumor markers in patients with meningeal carcinomatosis] No Shinkei Geka. 1991;19(12):1135–1141. [PubMed] [Google Scholar]
  • 46.Yoshida J, Yamamoto R, Wakabayashi T, et al. Radioimmunoassay of glioma-associated antigen in cerebrospinal fluid and its usefulness for the diagnosis and monitoring of human glioma. J Neurooncol. 1990;8(1):23–31. doi: 10.1007/BF00182083. [DOI] [PubMed] [Google Scholar]
  • 47.Prisell P, Persson L, Boethius J, et al. Somatomedins in tumour cyst fluid, cerebrospinal fluid, and tumour cytosol in patients with glial tumours. Acta Neurochir (Wien) 1987;89(1–2):48–52. doi: 10.1007/BF01406667. [DOI] [PubMed] [Google Scholar]
  • 48.Suzuki Y, Ishii R, Otsuka R, et al. [Clinical significance of beta 2-microglobulin and carcinoembryonic antigen in intracranial tumors] No To Shinkei. 1987;39(10):965–970. [PubMed] [Google Scholar]
  • 49.Gronowitz JS, Kallander CF, Hagberg H, et al. Deoxythymidine-kinase in cerebrospinal fluid: a new potential “marker” for brain tumours. Acta Neurochir (Wien) 1984;73(1–2):1–12. doi: 10.1007/BF01401779. [DOI] [PubMed] [Google Scholar]
  • 50.Suzuki Y, Tanaka R. Carcinoembryonic antigen in patients with intracranial tumors. J Neurosurg. 1980;53(3):355–360. doi: 10.3171/jns.1980.53.3.0355. [DOI] [PubMed] [Google Scholar]
  • 51.Miyake E, Yamashita M, Kitamura K, et al. Carcinoembryonic antigen (CEA) levels in patients with brain tumours. Acta Neurochir (Wien) 1979;46(1–2):53–57. doi: 10.1007/BF01407680. [DOI] [PubMed] [Google Scholar]
  • 52.Stott SL, Lee RJ, Nagrath S, et al. Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer. Sci Transl Med. 2010;2(25):25ra23. doi: 10.1126/scitranslmed.3000403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Muller V, Stahmann N, Riethdorf S, et al. Circulating tumor cells in breast cancer: correlation to bone marrow micrometastases, heterogeneous response to systemic therapy and low proliferative activity. Clin Cancer Res. 2005;11(10):3678–3685. doi: 10.1158/1078-0432.CCR-04-2469. [DOI] [PubMed] [Google Scholar]
  • 54.Pierga JY, Bidard FC, Mathiot C, et al. Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial. Clin Cancer Res. 2008;14(21):7004–7010. doi: 10.1158/1078-0432.CCR-08-0030. [DOI] [PubMed] [Google Scholar]
  • 55.Riethdorf S, Muller V, Zhang L, et al. Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin Cancer Res. 2010;16(9):2634–2645. doi: 10.1158/1078-0432.CCR-09-2042. [DOI] [PubMed] [Google Scholar]
  • 56.Xenidis N, Ignatiadis M, Apostolaki S, et al. Cytokeratin-19 mRNA-positive circulating tumor cells after adjuvant chemotherapy in patients with early breast cancer. J Clin Oncol. 2009;27(13):2177–2184. doi: 10.1200/JCO.2008.18.0497. [DOI] [PubMed] [Google Scholar]
  • 57.Li X, Wong C, Mysel R, et al. Screening and identification of differentially expressed transcripts in circulating cells of prostate cancer patients using suppression subtractive hybridization. Mol Cancer. 2005;4(1):30. doi: 10.1186/1476-4598-4-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pantel K, Alix-Panabieres C. Real-time liquid biopsy in cancer patients: fact or fiction? Cancer Res. 2013;73(21):6384–6388. doi: 10.1158/0008-5472.CAN-13-2030. [DOI] [PubMed] [Google Scholar]
  • 59.Zheng PP, Kros JM. Challenge of the gap between the current mania of cancer stem cells and the therapeutic strategy for patients with cancer. Int J Cancer. 2010;126(6):1529–1530. doi: 10.1002/ijc.24863. [DOI] [PubMed] [Google Scholar]
  • 60.Nagrath S, Sequist LV, Maheswaran S, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450(7173):1235–1239. doi: 10.1038/nature06385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Alix-Panabieres C, Pierga JY. [Circulating tumor cells: liquid biopsy] Cellules tumorales circulantes: biopsie liquide du cancer. Bull Cancer. 2014;101(1):17–23. doi: 10.1684/bdc.2014.1883. [DOI] [PubMed] [Google Scholar]
  • 62.Friedlander TW, Premasekharan G, Paris PL. Looking back, to the future of circulating tumor cells. Pharmacol Ther. 2014;142(3):271–280. doi: 10.1016/j.pharmthera.2013.12.011. [DOI] [PubMed] [Google Scholar]
  • 63.Lowes LE, Allan AL. Recent advances in the molecular characterization of circulating tumor cells. Cancers (Basel) 2014;6(1):595–624. doi: 10.3390/cancers6010595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Macarthur KM, Kao GD, Chandrasekaran S, et al. Detection of brain tumor cells in the peripheral blood by a telomerase promoter-based assay. Cancer Res. 2014;74(8):2152–2159. doi: 10.1158/0008-5472.CAN-13-0813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Luzzi KJ, MacDonald IC, Schmidt EE, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153(3):865–873. doi: 10.1016/S0002-9440(10)65628-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tarin D, Price JE, Kettlewell MG, et al. Mechanisms of human tumor metastasis studied in patients with peritoneovenous shunts. Cancer Res. 1984;44(8):3584–3592. [PubMed] [Google Scholar]
  • 67.Weiss L. Metastatic inefficiency. Adv Cancer Res. 1990;54:159–211. doi: 10.1016/s0065-230x(08)60811-8. [DOI] [PubMed] [Google Scholar]
  • 68.Heitzer E, Auer M, Gasch C, et al. Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing. Cancer Res. 2013;73(10):2965–2975. doi: 10.1158/0008-5472.CAN-12-4140. [DOI] [PubMed] [Google Scholar]
  • 69.Krebs MG, Metcalf RL, Carter L, et al. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat Rev Clin Oncol. 2014;11(3):129–144. doi: 10.1038/nrclinonc.2013.253. [DOI] [PubMed] [Google Scholar]
  • 70.Amitendu S, Mak SK, Ling JM, et al. A single institution experience of the incidence of extracranial metastasis in glioma. J Clin Neurosci. 2012;19(11):1511–1515. doi: 10.1016/j.jocn.2011.08.040. [DOI] [PubMed] [Google Scholar]
  • 71.Armstrong TS, Prabhu S, Aldape K, et al. A case of soft tissue metastasis from glioblastoma and review of the literature. J Neurooncol. 2011;103(1):167–172. doi: 10.1007/s11060-010-0370-y. [DOI] [PubMed] [Google Scholar]
  • 72.Blume C, von Lehe M, van Landeghem F, et al. Extracranial glioblastoma with synchronous metastases in the lung, pulmonary lymph nodes, vertebrae, cervical muscles and epidural space in a young patient - case report and review of literature. BMC Res Notes. 2013;6:290. doi: 10.1186/1756-0500-6-290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Can B, Akpolat I, Meydan D, et al. Fine-needle aspiration cytology of metastatic oligodendroglioma: case report and literature review. Acta Cytol. 2012;56(1):97–103. doi: 10.1159/000331769. [DOI] [PubMed] [Google Scholar]
  • 74.Dawar R, Fabiano AJ, Qiu J, et al. Secondary gliosarcoma with extra-cranial metastases: a report and review of the literature. Clin Neurol Neurosurg. 2013;115(4):375–380. doi: 10.1016/j.clineuro.2012.06.017. [DOI] [PubMed] [Google Scholar]
  • 75.Frank S, Kuhn SA, Brodhun M, et al. Metastatic glioblastoma cells use common pathways via blood and lymphatic vessels. Neurol Neurochir Pol. 2009;43(2):183–190. [PubMed] [Google Scholar]
  • 76.Kalokhe G, Grimm SA, Chandler JP, et al. Metastatic glioblastoma: case presentations and a review of the literature. J Neurooncol. 2012;107(1):21–27. doi: 10.1007/s11060-011-0731-1. [DOI] [PubMed] [Google Scholar]
  • 77.Li G, Zhang Z, Zhang J, et al. Occipital anaplastic oligodendroglioma with multiple organ metastases after a short clinical course: a case report and literature review. Diagn Pathol. 2014;9:17. doi: 10.1186/1746-1596-9-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Martens T, Matschke J, Muller C, et al. Skeletal spread of an anaplastic astrocytoma (WHO grade III) and preservation of histopathological properties within metastases. Clin Neurol Neurosurg. 2013;115(3):323–328. doi: 10.1016/j.clineuro.2012.05.025. [DOI] [PubMed] [Google Scholar]
  • 79.Saad AG, Sachs J, Turner CD, et al. Extracranial metastases of glioblastoma in a child: case report and review of the literature. J Pediatr Hematol Oncol. 2007;29(3):190–194. doi: 10.1097/MPH.0b013e31803350a7. [DOI] [PubMed] [Google Scholar]
  • 80.Takanen S, Bangrazi C, Caiazzo R, et al. Multiple bone metastases from glioblastoma multiforme without local brain relapse: a case report and review of the literature. Tumori. 2013;99(5):e237–e240. doi: 10.1177/030089161309900521. [DOI] [PubMed] [Google Scholar]
  • 81.Wu Y, Liu B, Qu L, et al. Extracranial skeletal metastasis in anaplastic oligodendroglioma: case report and review of the literature. J Int Med Res. 2011;39(3):960–967. doi: 10.1177/147323001103900331. [DOI] [PubMed] [Google Scholar]
  • 82.Armanios MY, Grossman SA, Yang SC, et al. Transmission of glioblastoma multiforme following bilateral lung transplantation from an affected donor: case study and review of the literature. Neuro Oncol. 2004;6(3):259–263. doi: 10.1215/S1152851703000474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Chen F, Karolak W, Cypel M, et al. Intermediate-term outcome in lung transplantation from a donor with glioblastoma multiforme. J Heart Lung Transplant. 2009;28(10):1116–1118. doi: 10.1016/j.healun.2009.06.022. [DOI] [PubMed] [Google Scholar]
  • 84.Chen H, Shah AS, Girgis RE, et al. Transmission of glioblastoma multiforme after bilateral lung transplantation. J Clin Oncol. 2008;26(19):3284–3285. doi: 10.1200/JCO.2008.16.3543. [DOI] [PubMed] [Google Scholar]
  • 85.Fatt MA, Horton KM, Fishman EK. Transmission of metastatic glioblastoma multiforme from donor to lung transplant recipient. J Comput Assist Tomogr. 2008;32(3):407–409. doi: 10.1097/RCT.0b013e318076b472. [DOI] [PubMed] [Google Scholar]
  • 86.Frank S, Muller J, Bonk C, et al. Transmission of glioblastoma multiforme through liver transplantation. Lancet. 1998;352(9121):31. doi: 10.1016/S0140-6736(98)24027-X. [DOI] [PubMed] [Google Scholar]
  • 87.Jonas S, Bechstein WO, Lemmens HP, et al. Liver graft-transmitted glioblastoma multiforme. A case report and experience with 13 multiorgan donors suffering from primary cerebral neoplasia. Transpl Int. 1996;9(4):426–429. doi: 10.1007/BF00335707. [DOI] [PubMed] [Google Scholar]
  • 88.Nauen DW, Li QK. Cytological diagnosis of metastatic glioblastoma in the pleural effusion of a lung transplant patient. Diagn Cytopathol. 2013;42(7):619–623. doi: 10.1002/dc.22993. [DOI] [PubMed] [Google Scholar]
  • 89.Val-Bernal F, Ruiz JC, Cotorruelo JG, et al. Glioblastoma multiforme of donor origin after renal transplantation: report of a case. Hum Pathol. 1993;24(11):1256–1259. doi: 10.1016/0046-8177(93)90224-5. [DOI] [PubMed] [Google Scholar]
  • 90.Cohen J, Dunbar E, Reynolds B. Circulating Tumor Cells in Patients with Glioblastoma (MeetingAbstracts.P06.006) Neurology. 2012 doi: 10.1212/WNL.78.1_MeetingAbstracts.P06.006Neurology. April 26, 2012; 78(Meeting Abstracts 1): P06.006. [DOI] [Google Scholar]
  • 91.Al-Nedawi K, Meehan B, Rak J. Microvesicles: messengers and mediators of tumor progression. Cell Cycle. 2009;8(13):2014–2018. doi: 10.4161/cc.8.13.8988. [DOI] [PubMed] [Google Scholar]
  • 92.D'Asti E, Garnier D, Lee TH, et al. Oncogenic extracellular vesicles in brain tumor progression. Front Physiol. 2012;3:294. doi: 10.3389/fphys.2012.00294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Taylor DD, Gercel-Taylor C. The origin, function, and diagnostic potential of RNA within extracellular vesicles present in human biological fluids. Front Genet. 2013;4:142. doi: 10.3389/fgene.2013.00142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Antonyak MA, Li B, Boroughs LK, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci USA. 2011;108(12):4852–4857. doi: 10.1073/pnas.1017667108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chen X, Liang H, Zhang J, et al. Secreted microRNAs: a new form of intercellular communication. Trends Cell Biol. 2012;22(3):125–132. doi: 10.1016/j.tcb.2011.12.001. [DOI] [PubMed] [Google Scholar]
  • 96.Skog J, Wurdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–1476. doi: 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Redzic JS, Ung TH, Graner MW. Glioblastoma extracellular vesicles: reservoirs of potential biomarkers. Pharmgenomics Pers Med. 2014;7:65–77. doi: 10.2147/PGPM.S39768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–579. doi: 10.1038/nri855. [DOI] [PubMed] [Google Scholar]
  • 99.Shao H, Chung J, Balaj L, et al. Protein typing of circulating microvesicles allows real-time monitoring of glioblastoma therapy. Nat Med. 2012;18(12):1835–1840. doi: 10.1038/nm.2994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Akers JC, Ramakrishnan V, Kim R, et al. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development. PLoS One. 2013;8(10):e78115. doi: 10.1371/journal.pone.0078115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kroeger KM, Muhammad AK, Baker GJ, et al. Gene therapy and virotherapy: novel therapeutic approaches for brain tumors. Discov Med. 2010;10(53):293–304. [PMC free article] [PubMed] [Google Scholar]
  • 102.Andrews DW. Pilot Immunotherapy Trial for Recurrent Malignant Gliomas. ClinicalTrials.gov. 2013 http://clinicaltrials.gov/show/NCT01550523 . [Google Scholar]
  • 103.Yoshioka Y, Kosaka N, Konishi Y, et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen. Nat Commun. 2014;5:3591. doi: 10.1038/ncomms4591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Witwer KW, Buzas EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2 doi: 10.3402/jev.v2i0.20360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426–437. doi: 10.1038/nrc3066. [DOI] [PubMed] [Google Scholar]
  • 106.Tumilson CA, Lea RW, Alder JE, et al. Circulating MicroRNA Biomarkers for Glioma and Predicting Response to Therapy. Mol Neurobiol. 2014 doi: 10.1007/s12035-014-8679-8.. [DOI] [PubMed] [Google Scholar]
  • 107.Diaz LA, Jr., Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32(6):579–586. doi: 10.1200/JCO.2012.45.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Boisselier B, Gallego Perez-Larraya J, Rossetto M, et al. Detection of IDH1 mutation in the plasma of patients with glioma. Neurology. 2012;79(16):1693–1698. doi: 10.1212/WNL.0b013e31826e9b0a. [DOI] [PubMed] [Google Scholar]
  • 109.Salkeni MA, Zarzour A, Ansay TY, et al. Detection of EGFRvIII mutant DNA in the peripheral blood of brain tumor patients. J Neurooncol. 2013;115(1):27–35. doi: 10.1007/s11060-013-1209-0. [DOI] [PubMed] [Google Scholar]
  • 110.Balana C, Ramirez JL, Taron M, et al. O6-methyl-guanine-DNA methyltransferase methylation in serum and tumor DNA predicts response to 1,3-bis(2-chloroethyl)-1-nitrosourea but not to temozolamide plus cisplatin in glioblastoma multiforme. Clin Cancer Res. 2003;9(4):1461–1468. [PubMed] [Google Scholar]
  • 111.Weaver KD, Grossman SA, Herman JG. Methylated tumor-specific DNA as a plasma biomarker in patients with glioma. Cancer Invest. 2006;24(1):35–40. doi: 10.1080/07357900500449546. [DOI] [PubMed] [Google Scholar]
  • 112.Ramirez JL, Taron M, Balana C, et al. Serum DNA as a tool for cancer patient management. Rocz Akad Med Bialymst. 2003;48:34–41. [PubMed] [Google Scholar]
  • 113.Wakabayashi T, Natsume A, Hatano H, et al. p16 promoter methylation in the serum as a basis for the molecular diagnosis of gliomas. Neurosurgery. 2009;64(3):455–461. doi: 10.1227/01.NEU.0000340683.19920.E3. discussion 461–452. [DOI] [PubMed] [Google Scholar]
  • 114.Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007;1775(1):181–232. doi: 10.1016/j.bbcan.2006.10.001. [DOI] [PubMed] [Google Scholar]
  • 115.Reddi KK, Holland JF. Elevated serum ribonuclease in patients with pancreatic cancer. Proc Natl Acad Sci USA. 1976;73(7):2308–2310. doi: 10.1073/pnas.73.7.2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Noerholm M, Balaj L, Limperg T, et al. RNA expression patterns in serum microvesicles from patients with glioblastoma multiforme and controls. BMC Cancer. 2012;12:22. doi: 10.1186/1471-2407-12-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jaiswal R, Luk F, Gong J, et al. Microparticle conferred microRNA profiles--implications in the transfer and dominance of cancer traits. Mol Cancer. 2012;11:37. doi: 10.1186/1476-4598-11-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Nilsson RJ, Balaj L, Hulleman E, et al. Blood platelets contain tumor-derived RNA biomarkers. Blood. 2011;118(13):3680–3683. doi: 10.1182/blood-2011-03-344408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6(4):259–269. doi: 10.1038/nrc1840. [DOI] [PubMed] [Google Scholar]
  • 120.Schwarzenbach H, Nishida N, Calin GA, et al. Clinical relevance of circulating cell-free microRNAs in cancer. Nat Rev Clin Oncol. 2014;11(3):145–156. doi: 10.1038/nrclinonc.2014.5. [DOI] [PubMed] [Google Scholar]
  • 121.Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6(11):857–866. doi: 10.1038/nrc1997. [DOI] [PubMed] [Google Scholar]
  • 122.Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65(14):6029–6033. doi: 10.1158/0008-5472.CAN-05-0137. [DOI] [PubMed] [Google Scholar]
  • 123.Haapa-Paananen S, Chen P, Hellstrom K, et al. Functional profiling of precursor MicroRNAs identifies MicroRNAs essential for glioma proliferation. PLoS One. 2013;8(4):e60930. doi: 10.1371/journal.pone.0060930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Jarry J, Schadendorf D, Greenwood C, et al. The validity of circulating microRNAs in oncology: Five years of challenges and contradictions. Mol Oncol. 2014;8(4):819–829. doi: 10.1016/j.molonc.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Malzkorn B, Wolter M, Liesenberg F, et al. Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol. 2010;20(3):539–550. doi: 10.1111/j.1750-3639.2009.00328.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rao SA, Santosh V, Somasundaram K. Genome-wide expression profiling identifies deregulated miRNAs in malignant astrocytoma. Mod Pathol. 2010;23(10):1404–1417. doi: 10.1038/modpathol.2010.135. [DOI] [PubMed] [Google Scholar]
  • 127.Silber J, Lim DA, Petritsch C, et al. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 2008;6:14. doi: 10.1186/1741-7015-6-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wang J, Zhang KY, Liu SM, et al. Tumor-associated circulating microRNAs as biomarkers of cancer. Molecules. 2014;19(2):1912–1938. doi: 10.3390/molecules19021912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wu L, Li G, Feng D, et al. MicroRNA-21 expression is associated with overall survival in patients with glioma. Diagn Pathol. 2013;8:200. doi: 10.1186/1746-1596-8-200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Teplyuk NM, Mollenhauer B, Gabriely G, et al. MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity. Neuro Oncol. 2012;14(6):689–700. doi: 10.1093/neuonc/nos074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Yang C, Wang C, Chen X, et al. Identification of seven serum microRNAs from a genome-wide serum microRNA expression profile as potential noninvasive biomarkers for malignant astrocytomas. Int J Cancer. 2013;132(1):116–127. doi: 10.1002/ijc.27657. [DOI] [PubMed] [Google Scholar]
  • 132.Baraniskin A, Kuhnhenn J, Schlegel U, et al. Identification of microRNAs in the cerebrospinal fluid as biomarker for the diagnosis of glioma. Neuro Oncol. 2012;14(1):29–33. doi: 10.1093/neuonc/nor169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Ilhan-Mutlu A, Wagner L, Wohrer A, et al. Blood alterations preceding clinical manifestation of glioblastoma. Cancer Invest. 2012;30(9):625–629. doi: 10.3109/07357907.2012.725443. [DOI] [PubMed] [Google Scholar]
  • 134.Ilhan-Mutlu A, Wagner L, Wohrer A, et al. Plasma MicroRNA-21 concentration may be a useful biomarker in glioblastoma patients. Cancer Invest. 2012;30(8):615–621. doi: 10.3109/07357907.2012.708071. [DOI] [PubMed] [Google Scholar]
  • 135.Wang Q, Li P, Li A, et al. Plasma specific miRNAs as predictive biomarkers for diagnosis and prognosis of glioma. J Exp Clin Cancer Res. 2012;31:97. doi: 10.1186/1756-9966-31-97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Roth P, Wischhusen J, Happold C, et al. A specific miRNA signature in the peripheral blood of glioblastoma patients. J Neurochem. 2011;118(3):449–457. doi: 10.1111/j.1471-4159.2011.07307.x. [DOI] [PubMed] [Google Scholar]
  • 137.Holdhoff M, Yovino SG, Boadu O, et al. Blood-based biomarkers for malignant gliomas. J Neurooncol. 2013;113(3):345–352. doi: 10.1007/s11060-013-1144-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Fine HA, Figg WD, Jaeckle K, et al. Phase II trial of the antiangiogenic agent thalidomide in patients with recurrent high-grade gliomas. J Clin Oncol. 2000;18(4):708–715. doi: 10.1200/JCO.2000.18.4.708. [DOI] [PubMed] [Google Scholar]
  • 139.Nakagawa H, Yamada M, Kanayama T, et al. Myelin basic protein in the cerebrospinal fluid of patients with brain tumors. Neurosurgery. 1994;34(5):825–833. doi: 10.1227/00006123-199405000-00006. discussion 833. [DOI] [PubMed] [Google Scholar]
  • 140.Onodera Y, Saitoh Y, Nagai K. [The diagnostic value of immunoglobulin G in the cerebrospinal fluid of brain tumor patients, particularly in malignant tumors] Gan No Rinsho. 1987;33(12):1402–1406. [PubMed] [Google Scholar]
  • 141.Rombos A, Evangelopoulu-Katsiri E, Mariatos P, et al. Cerebrospinal fluid carcinoembryonic antigen and alphafetoprotein in patients with central nervous system neoplasia. Acta Neurol Scand. 1988;77(6):440–444. doi: 10.1111/j.1600-0404.1988.tb05937.x. [DOI] [PubMed] [Google Scholar]
  • 142.Ransohoff J, Weiss J. Cerebrospinal fluid sterols in the evaluation of patients with gliomas. Natl Cancer Inst Monogr. 1977;46:119–124. [PubMed] [Google Scholar]
  • 143.Jung CS, Foerch C, Schanzer A, et al. Serum GFAP is a diagnostic marker for glioblastoma multiforme. Brain. 2007;130(Pt 12):3336–3341. doi: 10.1093/brain/awm263. [DOI] [PubMed] [Google Scholar]
  • 144.Takaue Y, Nishioka K, van Eys J. Evaluation of polyamine levels in cerebrospinal fluid of children with brain tumors. J Neurooncol. 1986;3(4):327–333. doi: 10.1007/BF00165581. [DOI] [PubMed] [Google Scholar]
  • 145.Zalatimo O, Zoccoli CM, Patel A, et al. Impact of genetic targets on primary brain tumor therapy: what's ready for prime time? Adv Exp Med Biol. 2013;779:267–289. doi: 10.1007/978-1-4614-6176-0_12. [DOI] [PubMed] [Google Scholar]
  • 146.Muller HL, Oh Y, Lehrnbecher T, et al. Insulin-like growth factor-binding protein-2 concentrations in cerebrospinal fluid and serum of children with malignant solid tumors or acute leukemia. J Clin Endocrinol Metab. 1994;79(2):428–434. doi: 10.1210/jcem.79.2.7519190. [DOI] [PubMed] [Google Scholar]
  • 147.Edwards MS, Hudgins RJ, Wilson CB, et al. Pineal region tumors in children. J Neurosurg. 1988;68(5):689–697. doi: 10.3171/jns.1988.68.5.0689. [DOI] [PubMed] [Google Scholar]
  • 148.Shinomiya Y, Toya S, Iwata T, et al. Radioimmunoassay of alpha-fetoprotein in children with primary intracranial tumors. Childs Brain. 1979;5(5):450–458. doi: 10.1159/000119840. [DOI] [PubMed] [Google Scholar]
  • 149.Li Y, Jiang T, Zhang J, et al. Elevated serum antibodies against insulin-like growth factor-binding protein-2 allow detecting early-stage cancers: evidences from glioma and colorectal carcinoma studies. Ann Oncol. 2012;23(9):2415–2422. doi: 10.1093/annonc/mds007. [DOI] [PubMed] [Google Scholar]
  • 150.Chinnaiyan P, Chowdhary S, Potthast L, et al. Phase I trial of vorinostat combined with bevacizumab and CPT-11 in recurrent glioblastoma. Neuro Oncol. 2012;14(1):93–100. doi: 10.1093/neuonc/nor187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hua C, Wu S, Chemaitilly W, et al. Predicting the probability of abnormal stimulated growth hormone response in children after radiotherapy for brain tumors. Int J Radiat Oncol Biol Phys. 2012;84(4):990–995. doi: 10.1016/j.ijrobp.2012.01.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Lonn S, Inskip PD, Pollak MN, et al. Glioma risk in relation to serum levels of insulin-like growth factors. Cancer Epidemiol Biomarkers Prev. 2007;16(4):844–846. doi: 10.1158/1055-9965.EPI-06-1010. [DOI] [PubMed] [Google Scholar]
  • 153.Weinzimer SA, Homan SA, Ferry RJ, et al. Serum IGF-I and IGFBP-3 concentrations do not accurately predict growth hormone deficiency in children with brain tumours. Clin Endocrinol (Oxf) 1999;51(3):339–345. doi: 10.1046/j.1365-2265.1999.00804.x. [DOI] [PubMed] [Google Scholar]
  • 154.Hwang SL, Lin CL, Cheng CY, et al. Serum concentration of soluble decoy receptor 3 in glioma patients before and after surgery. Kaohsiung J Med Sci. 2004;20(3):124–127. doi: 10.1016/S1607-551X(09)70095-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Zheng PP, Hop WC, Luider TM, et al. Increased levels of circulating endothelial progenitor cells and circulating endothelial nitric oxide synthase in patients with gliomas. Ann Neurol. 2007;62(1):40–48. doi: 10.1002/ana.21151. [DOI] [PubMed] [Google Scholar]
  • 156.Ijsselstijn L, Dekker LJ, Stingl C, et al. Serum levels of pregnancy zone protein are elevated in presymptomatic Alzheimer's disease. J Proteome Res. 2011;10(11):4902–4910. doi: 10.1021/pr200270z. [DOI] [PubMed] [Google Scholar]
  • 157.Kalinina J, Peng J, Ritchie JC, et al. Proteomics of gliomas: initial biomarker discovery and evolution of technology. Neuro Oncol. 2011;13(9):926–942. doi: 10.1093/neuonc/nor078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Liu J, Zheng S, Yu JK, et al. Serum protein fingerprinting coupled with artificial neural network distinguishes glioma from healthy population or brain benign tumor. J Zhejiang Univ Sci B. 2005;6(1):4–10. doi: 10.1631/jzus.2005.B0004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Zhang H, Wu G, Tu H, et al. Discovery of serum biomarkers in astrocytoma by SELDI-TOF MS and proteinchip technology. J Neurooncol. 2007;84(3):315–323. doi: 10.1007/s11060-007-9376-5. [DOI] [PubMed] [Google Scholar]
  • 160.Li J, Zhuang Z, Okamoto H, et al. Proteomic profiling distinguishes astrocytomas and identifies differential tumor markers. Neurology. 2006;66(5):733–736. doi: 10.1212/01.wnl.0000201270.90502.d0. [DOI] [PubMed] [Google Scholar]
  • 161.Saratsis AM, Yadavilli S, Magge S, et al. Insights into pediatric diffuse intrinsic pontine glioma through proteomic analysis of cerebrospinal fluid. Neuro Oncol. 2012;14(5):547–560. doi: 10.1093/neuonc/nos067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Rajcevic U, Petersen K, Knol JC, et al. iTRAQ-based proteomics profiling reveals increased metabolic activity and cellular cross-talk in angiogenic compared with invasive glioblastoma phenotype. Mol Cell Proteomics. 2009;8(11):2595–2612. doi: 10.1074/mcp.M900124-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Hoeben A, Landuyt B, Highley MS, et al. Vascular endothelial growth factor and angiogenesis. Pharmacol Rev. 2004;56(4):549–580. doi: 10.1124/pr.56.4.3. [DOI] [PubMed] [Google Scholar]
  • 164.Rijken PF, Bernsen HJ, Peters JP, et al. Spatial relationship between hypoxia and the (perfused) vascular network in a human glioma xenograft: a quantitative multi-parameter analysis. Int J Radiat Oncol Biol Phys. 2000;48(2):571–582. doi: 10.1016/s0360-3016(00)00686-6. [DOI] [PubMed] [Google Scholar]
  • 165.Soda Y, Myskiw C, Rommel A, et al. Mechanisms of neovascularization and resistance to anti-angiogenic therapies in glioblastoma multiforme. J Mol Med (Berl) 2013;91(4):439–448. doi: 10.1007/s00109-013-1019-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Norden AD, Drappatz J, Wen PY. Antiangiogenic therapies for high-grade glioma. Nat Rev Neurol. 2009;5(11):610–620. doi: 10.1038/nrneurol.2009.159. [DOI] [PubMed] [Google Scholar]
  • 167.Ferrara N, Hillan KJ, Novotny W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem Biophys Res Commun. 2005;333(2):328–335. doi: 10.1016/j.bbrc.2005.05.132. [DOI] [PubMed] [Google Scholar]
  • 168.Norden AD, Drappatz J, Wen PY. Novel anti-angiogenic therapies for malignant gliomas. Lancet Neurol. 2008;7(12):1152–1160. doi: 10.1016/S1474-4422(08)70260-6. [DOI] [PubMed] [Google Scholar]
  • 169.Gerstner ER, Batchelor TT. Antiangiogenic therapy for glioblastoma. Cancer J. 2012;18(1):45–50. doi: 10.1097/PPO.0b013e3182431c6f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Reardon DA, Conrad CA, Cloughesy T, et al. Phase I study of AEE788, a novel multitarget inhibitor of ErbB- and VEGF-receptor-family tyrosine kinases, in recurrent glioblastoma patients. Cancer Chemother Pharmacol. 2012;69(6):1507–1518. doi: 10.1007/s00280-012-1854-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27(28):4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 172.Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25(30):4722–4729. doi: 10.1200/JCO.2007.12.2440. [DOI] [PubMed] [Google Scholar]
  • 173.Munaut C, Noel A, Hougrand O, et al. Vascular endothelial growth factor expression correlates with matrix metalloproteinases MT1-MMP, MMP-2 and MMP-9 in human glioblastomas. Int J Cancer. 2003;106(6):848–855. doi: 10.1002/ijc.11313. [DOI] [PubMed] [Google Scholar]
  • 174.Tabatabai G, Stupp R. Primetime for antiangiogenic therapy. Curr Opin Neurol. 2009;22(6):639–644. doi: 10.1097/WCO.0b013e328332ba28. [DOI] [PubMed] [Google Scholar]
  • 175.Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8(8):592–603. doi: 10.1038/nrc2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Song S, Ewald AJ, Stallcup W, et al. PDGFRbeta+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol. 2005;7(9):870–879. doi: 10.1038/ncb1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kesari S, Schiff D, Henson JW, et al. Phase II study of temozolomide, thalidomide, and celecoxib for newly diagnosed glioblastoma in adults. Neuro Oncol. 2008;10(3):300–308. doi: 10.1215/15228517-2008-005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Rafat N, Beck G, Schulte J, et al. Circulating endothelial progenitor cells in malignant gliomas. J Neurosurg. 2010;112(1):43–49. doi: 10.3171/2009.5.JNS081074. [DOI] [PubMed] [Google Scholar]
  • 179.Salven P, Manpaa H, Orpana A, et al. Serum vascular endothelial growth factor is often elevated in disseminated cancer. Clin Cancer Res. 1997;3(5):647–651. [PubMed] [Google Scholar]
  • 180.Takano S, Yoshii Y, Kondo S, et al. Concentration of vascular endothelial growth factor in the serum and tumor tissue of brain tumor patients. Cancer Res. 1996;56(9):2185–2190. [PubMed] [Google Scholar]
  • 181.Schmidt NO, Westphal M, Hagel C, et al. Levels of vascular endothelial growth factor, hepatocyte growth factor/scatter factor and basic fibroblast growth factor in human gliomas and their relation to angiogenesis. Int J Cancer. 1999;84(1):10–18. doi: 10.1002/(sici)1097-0215(19990219)84:1<10::aid-ijc3>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
  • 182.Ilhan A, Gartner W, Neziri D, et al. Angiogenic factors in plasma of brain tumour patients. Anticancer Res. 2009;29(2):731–736. [PubMed] [Google Scholar]
  • 183.Berezov TT, Ovchinnikova LK, Kuznetsova OM, et al. Vascular endothelial growth factor in the serum of breast cancer patients. Bull Exp Biol Med. 2009;148(3):419–424. doi: 10.1007/s10517-010-0727-4. [DOI] [PubMed] [Google Scholar]
  • 184.Zhang SJ, Hu Y, Qian HL, et al. Expression and significance of ER, PR, VEGF, CA15–3, CA125 and CEA in judging the prognosis of breast cancer. Asian Pac J Cancer Prev. 2013;14(6):3937–3940. doi: 10.7314/apjcp.2013.14.6.3937. [DOI] [PubMed] [Google Scholar]
  • 185.Thielemann A, Baszczuk A, Kopczynski Z, et al. Clinical usefulness of assessing VEGF and soluble receptors sVEGFR-1 and sVEGFR-2 in women with breast cancer. Ann Agric Environ Med. 2013;20(2):293–297. [PubMed] [Google Scholar]
  • 186.Zhang Y, Meng X, Zeng H, et al. Serum vascular endothelial growth factor-C levels: A possible diagnostic marker for lymph node metastasis in patients with primary non-small cell lung cancer. Oncol Lett. 2013;6(2):545–549. doi: 10.3892/ol.2013.1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Liang J, Qian Y, Xu D, et al. Serum tumor markers, hypoxia-inducible factor-1alpha HIF-1alpha and vascular endothelial growth factor, in patients with non- small cell lung cancer before and after intervention. Asian Pac J Cancer Prev. 2013;14(6):3851–3854. doi: 10.7314/apjcp.2013.14.6.3851. [DOI] [PubMed] [Google Scholar]
  • 188.Sulkowska M, Famulski W, Wincewicz A, et al. Levels of VE-cadherin increase independently of VEGF in preoperative sera of patients with colorectal cancer. Tumori. 2006;92(1):67–71. doi: 10.1177/030089160609200111. [DOI] [PubMed] [Google Scholar]
  • 189.Sulkowski S, Wincewicz A, Zalewski B, et al. Hypoxia related growth factors and p53 in preoperative sera from patients with colorectal cancer--evaluation of the prognostic significance of these agents. Clin Chem Lab Med. 2009;47(11):1439–1445. doi: 10.1515/CCLM.2009.305. [DOI] [PubMed] [Google Scholar]
  • 190.Werther K, Christensen IJ, Brunner N, et al. Soluble vascular endothelial growth factor levels in patients with primary colorectal carcinoma. The Danish RANX05 Colorectal Cancer Study Group. Eur J Surg Oncol. 2000;26(7):657–662. doi: 10.1053/ejso.2000.0977. [DOI] [PubMed] [Google Scholar]
  • 191.Demirci U, Yaman M, Buyukberber S, et al. Prognostic importance of markers for inflammation, angiogenesis and apoptosis in high grade glial tumors during temozolomide and radiotherapy. Int Immunopharmacol. 2012;14(4):546–549. doi: 10.1016/j.intimp.2012.08.007. [DOI] [PubMed] [Google Scholar]
  • 192.Drevs J. Soluble markers for the detection of hypoxia under antiangiogenic treatment. Anticancer Res. 2003;23(2A):1159–1161. [PubMed] [Google Scholar]
  • 193.Batchelor TT, Gerstner ER, Emblem KE, et al. Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation. Proc Natl Acad Sci USA. 2013;110(47):19059–19064. doi: 10.1073/pnas.1318022110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Batchelor TT, Duda DG, di Tomaso E, et al. Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol. 2010;28(17):2817–2823. doi: 10.1200/JCO.2009.26.3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Lin Y, Jiang T, Zhou K, et al. Plasma IGFBP-2 levels predict clinical outcomes of patients with high-grade gliomas. Neuro Oncol. 2009;11(5):468–476. doi: 10.1215/15228517-2008-114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Fish JE, Marsden PA. Endothelial nitric oxide synthase: insight into cell-specific gene regulation in the vascular endothelium. Cell Mol Life Sci. 2006;63(2):144–162. doi: 10.1007/s00018-005-5421-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Duda DG, Fukumura D, Jain RK. Role of eNOS in neovascularization: NO for endothelial progenitor cells. Trends Mol Med. 2004;10(4):143–145. doi: 10.1016/j.molmed.2004.02.001. [DOI] [PubMed] [Google Scholar]
  • 198.Zheng PP, Hop WC, Sillevis Smitt PA, et al. Low-molecular weight caldesmon as a potential serum marker for glioma. Clin Cancer Res. 2005;11(12):4388–4392. doi: 10.1158/1078-0432.CCR-04-2512. [DOI] [PubMed] [Google Scholar]
  • 199.Zheng PP, Sieuwerts AM, Luider TM, et al. Differential expression of splicing variants of the human caldesmon gene (CALD1) in glioma neovascularization versus normal brain microvasculature. Am J Pathol. 2004;164(6):2217–2228. doi: 10.1016/S0002-9440(10)63778-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Zheng PP, van der Weiden M, Kros JM. Differential expression of Hela-type caldesmon in tumour neovascularization: a new marker of angiogenic endothelial cells. J Pathol. 2005;205(3):408–414. doi: 10.1002/path.1700. [DOI] [PubMed] [Google Scholar]
  • 201.Zheng PP, van der Weiden M, Kros JM. Hela l-CaD is implicated in the migration of endothelial cells/endothelial progenitor cells in human neoplasms. Cell Adh Migr. 2007;1(2):84–91. doi: 10.4161/cam.1.2.4332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Zheng PP, Weiden M, Sillevis Smitt PA, et al. Hela/-CaD undergoes a DNA replication-associated switch in localization from the cytoplasm to the nuclei of endothelial cells/endothelial progenitor cells in human tumor vasculature. Cancer Biol Ther. 2007;6(6):886–890. doi: 10.4161/cbt.6.6.4091. [DOI] [PubMed] [Google Scholar]
  • 203.Zheng PP, Severijnen LA, van der Weiden M, et al. A crucial role of caldesmon in vascular development in vivo. Cardiovasc Res. 2009;81(2):362–369. doi: 10.1093/cvr/cvn294. [DOI] [PubMed] [Google Scholar]
  • 204.Chong HC, Tan CK, Huang RL, et al. Matricellular proteins: a sticky affair with cancers. J Oncol. 2012;2012:351089. doi: 10.1155/2012/351089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Bornstein P. Matricellular proteins: an overview. J Cell Commun Signal. 2009;3(3–4):163–165. doi: 10.1007/s12079-009-0069-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Bornstein P. Diversity of function is inherent in matricellular proteins: an appraisal of thrombospondin 1. J Cell Biol. 1995;130(3):503–506. doi: 10.1083/jcb.130.3.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Sreekanthreddy P, Srinivasan H, Kumar DM, et al. Identification of potential serum biomarkers of glioblastoma: serum osteopontin levels correlate with poor prognosis. Cancer Epidemiol Biomarkers Prev. 2010;19(6):1409–1422. doi: 10.1158/1055-9965.EPI-09-1077. [DOI] [PubMed] [Google Scholar]
  • 208.El-Tanani MK, Campbell FC, Kurisetty V, et al. The regulation and role of osteopontin in malignant transformation and cancer. Cytokine Growth Factor Rev. 2006;17(6):463–474. doi: 10.1016/j.cytogfr.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 209.Minn AJ, Gupta GP, Siegel PM, et al. Genes that mediate breast cancer metastasis to lung. Nature. 2005;436(7050):518–524. doi: 10.1038/nature03799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347(25):1999–2009. doi: 10.1056/NEJMoa021967. [DOI] [PubMed] [Google Scholar]
  • 211.Kao CL, Chiou SH, Ho DM, et al. Elevation of plasma and cerebrospinal fluid osteopontin levels in patients with atypical teratoid/rhabdoid tumor. Am J Clin Pathol. 2005;123(2):297–304. doi: 10.1309/0ftkbkvnk4t5p1l1. [DOI] [PubMed] [Google Scholar]
  • 212.Nelson AR, Fingleton B, Rothenberg ML, et al. Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol. 2000;18(5):1135–1149. doi: 10.1200/JCO.2000.18.5.1135. [DOI] [PubMed] [Google Scholar]
  • 213.Borkakoti N. Structural studies of matrix metalloproteinases. J Mol Med (Berl) 2000;78(5):261–268. doi: 10.1007/s001090000113. [DOI] [PubMed] [Google Scholar]
  • 214.Forsyth PA, Wong H, Laing TD, et al. Gelatinase-A (MMP-2), gelatinase-B (MMP-9) and membrane type matrix metalloproteinase-1 (MT1-MMP) are involved in different aspects of the pathophysiology of malignant gliomas. Br J Cancer. 1999;79(11-12):1828–1835. doi: 10.1038/sj.bjc.6990291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Raithatha SA, Muzik H, Muzik H, et al. Localization of gelatinase-A and gelatinase-B mRNA and protein in human gliomas. Neuro Oncol. 2000;2(3):145–150. doi: 10.1093/neuonc/2.3.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Choe G, Park JK, Jouben-Steele L, et al. Active matrix metalloproteinase 9 expression is associated with primary glioblastoma subtype. Clin Cancer Res. 2002;8(9):2894–2901. [PubMed] [Google Scholar]
  • 217.Liu L, Wu J, Ying Z, et al. Astrocyte elevated gene-1 upregulates matrix metalloproteinase-9 and induces human glioma invasion. Cancer Res. 2010;70(9):3750–3759. doi: 10.1158/0008-5472.CAN-09-3838. [DOI] [PubMed] [Google Scholar]
  • 218.Iwamoto FM, Hottinger AF, Karimi S, et al. Longitudinal prospective study of matrix metalloproteinase-9 as a serum marker in gliomas. J Neurooncol. 2011;105(3):607–612. doi: 10.1007/s11060-011-0628-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Hormigo A, Gu B, Karimi S, et al. YKL-40 and matrix metalloproteinase-9 as potential serum biomarkers for patients with high-grade gliomas. Clin Cancer Res. 2006;12(19):5698–5704. doi: 10.1158/1078-0432.CCR-06-0181. [DOI] [PubMed] [Google Scholar]
  • 220.Wong ET, Alsop D, Lee D, et al. Cerebrospinal fluid matrix metalloproteinase-9 increases during treatment of recurrent malignant gliomas. Cerebrospinal Fluid Res. 2008;5:1. doi: 10.1186/1743-8454-5-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Friedberg MH, Glantz MJ, Klempner MS, et al. Specific matrix metalloproteinase profiles in the cerebrospinal fluid correlated with the presence of malignant astrocytomas, brain metastases, and carcinomatous meningitis. Cancer. 1998;82(5):923–930. doi: 10.1002/(sici)1097-0142(19980301)82:5<923::aid-cncr18>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 222.Vos CM, Gartner S, Ransohoff RM, et al. Matrix metalloprotease-9 release from monocytes increases as a function of differentiation: implications for neuroinflammation and neurodegeneration. J Neuroimmunol. 2000;109(2):221–227. doi: 10.1016/s0165-5728(00)00308-8. [DOI] [PubMed] [Google Scholar]
  • 223.Gijbels K, Masure S, Carton H, et al. Gelatinase in the cerebrospinal fluid of patients with multiple sclerosis and other inflammatory neurological disorders. J Neuroimmunol. 1992;41(1):29–34. doi: 10.1016/0165-5728(92)90192-n. [DOI] [PubMed] [Google Scholar]
  • 224.Rosenberg GA. Matrix metalloproteinases in brain injury. J Neurotrauma. 1995;12(5):833–842. doi: 10.1089/neu.1995.12.833. [DOI] [PubMed] [Google Scholar]
  • 225.Yadav L, Puri N, Rastogi V, et al. Matrix metalloproteinases and cancer - roles in threat and therapy. Asian Pac J Cancer Prev. 2014;15(3):1085–1091. doi: 10.7314/apjcp.2014.15.3.1085. [DOI] [PubMed] [Google Scholar]
  • 226.Brommeland T, Rosengren L, Fridlund S, et al. Serum levels of glial fibrillary acidic protein correlate to tumour volume of high-grade gliomas. Acta Neurol Scand. 2007;116(6):380–384. doi: 10.1111/j.1600-0404.2007.00889.x. [DOI] [PubMed] [Google Scholar]
  • 227.Lyubimova NV, Toms MG, Popova EE, et al. Neurospecific proteins in the serum of patients with brain tumors. Bull Exp Biol Med. 2011;150(6):732–734. doi: 10.1007/s10517-011-1236-9. [DOI] [PubMed] [Google Scholar]
  • 228.Wei P, Zhang W, Yang LS, et al. Serum GFAP autoantibody as an ELISA-detectable glioma marker. Tumour Biol. 2013;34(4):2283–2292. doi: 10.1007/s13277-013-0770-7. [DOI] [PubMed] [Google Scholar]
  • 229.Hammarstrom S. The carcinoembryonic antigen (CEA) family: structures, suggested functions and expression in normal and malignant tissues. Semin Cancer Biol. 1999;9(2):67–81. doi: 10.1006/scbi.1998.0119. [DOI] [PubMed] [Google Scholar]
  • 230.Rudenko VA, Lisianyi NI, Grinevich Iu A, et al. The level of embryonic antigens in the blood serum and cerebrospinal fluid of neuro-oncological patients. Zh Vopr Neirokhir Im N N Burdenko. 1996;(Jul–Sep(3)):32–35. [PubMed] [Google Scholar]
  • 231.Mitsios JV, McClellan A, Brown S, et al. Human chorionic gonadotropin and alpha-fetoprotein in cerebral spinal fluid: Method validation and retrospective review. Clin Biochem. 2014;47(7–8):632–635. doi: 10.1016/j.clinbiochem.2014.01.008. [DOI] [PubMed] [Google Scholar]
  • 232.Borger DR, Goyal L, Yau T, et al. Circulating oncometabolite 2-hydroxyglutarate is a potential surrogate biomarker in patients with isocitrate dehydrogenase-mutant intrahepatic cholangiocarcinoma. Clin Cancer Res. 2014;20(7):1884–1890. doi: 10.1158/1078-0432.CCR-13-2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.DiNardo CD, Propert KJ, Loren AW, et al. Serum 2-hydroxyglutarate levels predict isocitrate dehydrogenase mutations and clinical outcome in acute myeloid leukemia. Blood. 2013;121(24):4917–4924. doi: 10.1182/blood-2013-03-493197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Capper D, Simon M, Langhans CD, et al. 2-Hydroxyglutarate concentration in serum from patients with gliomas does not correlate with IDH1/2 mutation status or tumor size. Int J Cancer. 2012;131(3):766–768. doi: 10.1002/ijc.26425. [DOI] [PubMed] [Google Scholar]
  • 235.Bernardi D, Padoan A, Ballin A, et al. Serum YKL-40 following resection for cerebral glioblastoma. J Neurooncol. 2012;107(2):299–305. doi: 10.1007/s11060-011-0762-7. [DOI] [PubMed] [Google Scholar]
  • 236.Pelloski CE, Mahajan A, Maor M, et al. YKL-40 expression is associated with poorer response to radiation and shorter overall survival in glioblastoma. Clin Cancer Res. 2005;11(9):3326–3334. doi: 10.1158/1078-0432.CCR-04-1765. [DOI] [PubMed] [Google Scholar]
  • 237.Boot RG, van Achterberg TA, van Aken BE, et al. Strong induction of members of the chitinase family of proteins in atherosclerosis: chitotriosidase and human cartilage gp-39 expressed in lesion macrophages. Arterioscler Thromb Vasc Biol. 1999;19(3):687–694. doi: 10.1161/01.atv.19.3.687. [DOI] [PubMed] [Google Scholar]
  • 238.Clancy R. Nitric oxide alters chondrocyte function by disrupting cytoskeletal signaling complexes. Osteoarthritis Cartilage. 1999;7(4):399–400. doi: 10.1053/joca.1998.0223. [DOI] [PubMed] [Google Scholar]
  • 239.De Ceuninck F, Gaufillier S, Bonnaud A, et al. YKL-40 (cartilage gp-39) induces proliferative events in cultured chondrocytes and synoviocytes and increases glycosaminoglycan synthesis in chondrocytes. Biochem Biophys Res Commun. 2001;285(4):926–931. doi: 10.1006/bbrc.2001.5253. [DOI] [PubMed] [Google Scholar]
  • 240.Garnero P, Piperno M, Gineyts E, et al. Cross sectional evaluation of biochemical markers of bone, cartilage, and synovial tissue metabolism in patients with knee osteoarthritis: relations with disease activity and joint damage. Ann Rheum Dis. 2001;60(6):619–626. doi: 10.1136/ard.60.6.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Nordenbaek C, Johansen JS, Junker P, et al. YKL-40, a matrix protein of specific granules in neutrophils, is elevated in serum of patients with community-acquired pneumonia requiring hospitalization. J Infect Dis. 1999;180(5):1722–1726. doi: 10.1086/315050. [DOI] [PubMed] [Google Scholar]
  • 242.Volck B, Price PA, Johansen JS, et al. YKL-40, a mammalian member of the chitinase family, is a matrix protein of specific granules in human neutrophils. Proc Assoc Am Physicians. 1998;110(4):351–360. [PubMed] [Google Scholar]
  • 243.Biggar RJ, Johansen JS, Smedby KE, et al. Serum YKL-40 and interleukin 6 levels in Hodgkin lymphoma. Clin Cancer Res. 2008;14(21):6974–6978. doi: 10.1158/1078-0432.CCR-08-1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Johansen JS, Drivsholm L, Price PA, et al. High serum YKL-40 level in patients with small cell lung cancer is related to early death. Lung Cancer. 2004;46(3):333–340. doi: 10.1016/j.lungcan.2004.05.010. [DOI] [PubMed] [Google Scholar]
  • 245.Bergmann OJ, Johansen JS, Klausen TW, et al. High serum concentration of YKL-40 is associated with short survival in patients with acute myeloid leukemia. Clin Cancer Res. 2005;11(24 Pt 1):8644–8652. doi: 10.1158/1078-0432.CCR-05-1317. [DOI] [PubMed] [Google Scholar]
  • 246.Schmidt H, Johansen JS, Sjoegren P, et al. Serum YKL-40 predicts relapse-free and overall survival in patients with American Joint Committee on Cancer stage I and II melanoma. J Clin Oncol. 2006;24(5):798–804. doi: 10.1200/JCO.2005.03.7960. [DOI] [PubMed] [Google Scholar]
  • 247.Schmidt H, Johansen JS, Gehl J, et al. Elevated serum level of YKL-40 is an independent prognostic factor for poor survival in patients with metastatic melanoma. Cancer. 2006;106(5):1130–1139. doi: 10.1002/cncr.21678. [DOI] [PubMed] [Google Scholar]
  • 248.Cintin C, Johansen JS, Christensen IJ, et al. Serum YKL-40 and colorectal cancer. Br J Cancer. 1999;79(9–10):1494–1499. doi: 10.1038/sj.bjc.6690238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Johansen JS, Bojesen SE, Mylin AK, et al. Elevated plasma YKL-40 predicts increased risk of gastrointestinal cancer and decreased survival after any cancer diagnosis in the general population. J Clin Oncol. 2009;27(4):572–578. doi: 10.1200/JCO.2008.18.8367. [DOI] [PubMed] [Google Scholar]
  • 250.Hogdall EV, Johansen JS, Kjaer SK, et al. High plasma YKL-40 level in patients with ovarian cancer stage III is related to shorter survival. Oncol Rep. 2003;10(5):1535–1538. [PubMed] [Google Scholar]
  • 251.Dehn H, Hogdall EV, Johansen JS, et al. Plasma YKL-40, as a prognostic tumor marker in recurrent ovarian cancer. Acta Obstet Gynecol Scand. 2003;82(3):287–293. doi: 10.1034/j.1600-0412.2003.00010.x. [DOI] [PubMed] [Google Scholar]
  • 252.Dupont J, Tanwar MK, Thaler HT, et al. Early detection and prognosis of ovarian cancer using serum YKL-40. J Clin Oncol. 2004;22(16):3330–3339. doi: 10.1200/JCO.2004.09.112. [DOI] [PubMed] [Google Scholar]
  • 253.Johansen JS, Christensen IJ, Riisbro R, et al. High serum YKL-40 levels in patients with primary breast cancer is related to short recurrence free survival. Breast Cancer Res Treat. 2003;80(1):15–21. doi: 10.1023/A:1024431000710. [DOI] [PubMed] [Google Scholar]
  • 254.Jensen BV, Johansen JS, Price PA. High levels of serum HER-2/neu and YKL-40 independently reflect aggressiveness of metastatic breast cancer. Clin Cancer Res. 2003;9(12):4423–4434. [PubMed] [Google Scholar]
  • 255.Johansen JS, Brasso K, Iversen P, et al. Changes of biochemical markers of bone turnover and YKL-40 following hormonal treatment for metastatic prostate cancer are related to survival. Clin Cancer Res. 2007;13(11):3244–3249. doi: 10.1158/1078-0432.CCR-06-2616. [DOI] [PubMed] [Google Scholar]
  • 256.Tanwar MK, Gilbert MR, Holland EC. Gene expression microarray analysis reveals YKL-40 to be a potential serum marker for malignant character in human glioma. Cancer Res. 2002;62(15):4364–4368. [PubMed] [Google Scholar]
  • 257.Iwamoto FM, Hottinger AF, Karimi S, et al. Serum YKL-40 is a marker of prognosis and disease status in high-grade gliomas. Neuro Oncol. 2011;13(11):1244–1251. doi: 10.1093/neuonc/nor117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Verschuere T, Van Woensel M, Fieuws S, et al. Altered galectin-1 serum levels in patients diagnosed with high-grade glioma. J Neurooncol. 2013;115(1):9–17. doi: 10.1007/s11060-013-1201-8. [DOI] [PubMed] [Google Scholar]
  • 259.Ohnishi M, Matsumoto T, Nagashio R, et al. Proteomics of tumor-specific proteins in cerebrospinal fluid of patients with astrocytoma: usefulness of gelsolin protein. Pathol Int. 2009;59(11):797–803. doi: 10.1111/j.1440-1827.2009.02447.x. [DOI] [PubMed] [Google Scholar]
  • 260.Petrik V, Saadoun S, Loosemore A, et al. Serum alpha 2-HS glycoprotein predicts survival in patients with glioblastoma. Clin Chem. 2008;54(4):713–722. doi: 10.1373/clinchem.2007.096792. [DOI] [PubMed] [Google Scholar]
  • 261.Reddy PS, Umesh S, Thota B, et al. PBEF1/NAmPRTase/Visfatin: a potential malignant astrocytoma/glioblastoma serum marker with prognostic value. Cancer Biol Ther. 2008;7(5):663–668. doi: 10.4161/cbt.7.5.5663. [DOI] [PubMed] [Google Scholar]
  • 262.Iwadate Y, Hayama M, Adachi A, et al. High serum level of plasminogen activator inhibitor-1 predicts histological grade of intracerebral gliomas. Anticancer Res. 2008;28(1B):415–418. [PubMed] [Google Scholar]
  • 263.Todaro L, Christiansen S, Varela M, et al. Alteration of serum and tumoral neural cell adhesion molecule (NCAM) isoforms in patients with brain tumors. J Neurooncol. 2007;83(2):135–144. doi: 10.1007/s11060-006-9312-0. [DOI] [PubMed] [Google Scholar]
  • 264.Quaranta M, Divella R, Daniele A, et al. Epidermal growth factor receptor serum levels and prognostic value in malignant gliomas. Tumori. 2007;93(3):275–280. doi: 10.1177/030089160709300308. [DOI] [PubMed] [Google Scholar]
  • 265.Ilzecka J, Ilzecki M. APRIL is increased in serum of patients with brain glioblastoma multiforme. Eur Cytokine Netw. 2006;17(4):276–280. [PubMed] [Google Scholar]
  • 266.Fukuda ME, Iwadate Y, Machida T, et al. Cathepsin D is a potential serum marker for poor prognosis in glioma patients. Cancer Res. 2005;65(12):5190–5194. doi: 10.1158/0008-5472.CAN-04-4134. [DOI] [PubMed] [Google Scholar]
  • 267.Okada H, Yoshida J, Seo H, et al. Anti-(glioma surface antigen) monoclonal antibody G-22 recognizes overexpressed CD44 in glioma cells. Cancer Immunol Immunother. 1994;39(5):313–317. doi: 10.1007/BF01519984. [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