Abstract
Background:
Glioblastoma (GBM) is the most frequent and most malignant central nervous system (CNS) tumor. GBM shows poor prognosis with a median overall survival of 14.6 months, despite current surgical and adjuvant therapies. O(6)-methylguanine-DNA methyltransferase (MGMT) methylation is the strongest molecular prognosticator for GBM with therapeutic implications in adjuvant treatment. Isocitrate dehydrogenase (IDH) mutation is the most recently introduced molecular marker and is important for the GBM classification because distinguishes primary (de novo) from secondary GBM. In the last two decades huge advances in the understanding of biopathological bases of gliomagenesis have been made but, to date, there is a lack of biopathological markers endowed of some prognostic and predictive value for GBM.
Aim:
In the present review we analyzed the role, as possible prognosticators, of epidermal growth factor receptor (EGFR) variant III (EGFRvIII), phosphatase and tensin homolog (PTEN) deletion and other alteration of the receptor tyrosine kinase (RTK) pathway, and vascular endothelial growth factor (VEGF) expression. We included in the review studies considering both the prognostic value and the predictive value for response to therapy of the above-mentioned biomarkers.
Relevance for patients:
These factors have a paramount importance in gliomagenesis and are potential targets for individualized therapies. EGFR can be targeted by tyrosine kinase inhibitors (TKIs). mTOR, whose activation is triggered by PTEN loss, is the target of rapalogs and VEGF is the target of the molecular antibody bevacizumab. Unfortunately, current evidence is insufficient to draw a definite prognostic/predictive role for these biomarkers in GBM. Further understanding the gliomagenesis pathways and looking for biomarkers endowed with translational relevance are necessary efforts in order to find the appropriate, tailored therapy for each specific GBM patient.
Keywords: glioblastoma multiforme, prognosis, molecular marker, survival, O(6)-methylguanine-DNA methyltransferase, epidermal growth factor receptor, epidermal growth factor receptor variant III, phosphatase and tensin homolog deletion, vascular endothelial growth factor
1. Introduction
Glioblastoma (GBM) is the most frequent and most malignant central nervous system (CNS) tumor [1]. In the USA, GBM incidence is 3.9 cases/100000/year, representing 65-70% of astrocytic tumors and 12-15% of CNS neoplasms with a classical incidence peak between 55 and 74 years [2]. Prognosis of GBM is currently dismal, with a median survival from diagnosis of 14.6 months [3]. Standard-of-care is surgery, intended as maximal safe resection [4], followed by conformal radiotherapy and temozolomide (TMZ) given concurrently to radiotherapy and then as adjuvant therapy for at least 6 cycles [3]. Recent studies evaluating the role of other drugs, particularly bevacizumab, in the management of GBM, failed to establish a new standard-of-care [5,6]. Moreover, upon recurrence, no evidence on the best treatment choice exists [7,8]. The 2007 World Health Organization (WHO) classification of CNS tumors [1] failed to incorporate molecular markers for GBM classification purposes. The upcoming 2016 WHO classification of CNS tumors will likely take into account molecular prognosticators in a renowned “integrated diagnosis” [9]. In detail, the roles of O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation and isocitrate dehydrogenase (IDH) mutation are expected to be considered for GBM.
MGMT methylation is currently the strongest molecular prognosticator for GBM [10], though some doubts on its reliability in the first 6 months after GBM diagnosis have been raised [11]. MGMT promoter methylation carries therapeutic implications in adjuvant treatment of GBM in elderly [4]. IDH mutation is the most recently introduced molecular marker for astrocytic tumors, and is considered the first event in multistep gliomagenesis [12]. Therefore, IDH mutation distinguishes primary (de novo) from secondary GBM [13].
In the last two decades huge advances in the understanding of biopathological bases of gliomagenesis have been made and have unveiled a high grade of variability among different GBMs and also among different zones of the same tumor. Thus, GBM is considered as the archetype of heterogeneous cancer [13]. Nonetheless, despite the improvement of knowledge of gliomagenesis, there is a lack of biopathological markers endowed of some prognostic and predictive value. The ideal biomarker in GBM should be easy detectable by routine pathological techniques and highly reproducible among different laboratories and observers. Immunohistochemistry is a reliable, quick, no expensive and widely available technique, but sometimes the more accurate semiquantitative RT-PCR should be preferred. Moreover, an ideal biomarker should clearly identify patients with longer or shorter survival (prognostic role) and/or patients that can benefit from a particular treatment (predictive role). The above-mentioned markers (MGMT and IDH) have an established prognostic/predictive role in GBM and will not be discussed in further detail.
In the present review we will analyze the role as possible prognosticators, of epidermal growth factor receptor (EGFR) variant III (EGFRvIII), phosphatase and tensin homolog (PTEN) deletion and other alteration of the receptor tyrosine kinase (RTK) pathway, and vascular endothelial growth factor (VEGF) expression. At our Institution, EGFRvIII expression, PTEN/mTOR status and VEGF expression have been routinely studied for a decade [8,14,15] not only because they have a paramount importance in gliomagenesis, but also for their role as potential targets for individualized therapies. More specifically, EGFR can be targeted by tyrosine kinase inhibitors (TKIs). It has been showed that EGFRvIII expression is a predictive biomarker for response to TKIs, particularly erlotinib [8,16]. mTOR, whose activation is triggered by PTEN loss, is the target of rapalogs; finally, VEGF is the target of the molecular antibody bevacizumab[5,6]. There has been much enthusiasm in the past years regarding these “targeted therapies”, but results of clinical trials have been rather disappointing. Currently, bevacizumab remains a therapeutic option in recurrent GBM in the USA, whereas erlotinib and rapalogs are experimental drugs whose effectiveness has not been confirmed in unselected cohorts of patients. These evidences foster the search for biomarkers that can identify subgroups of patients who can benefit from targeted therapies [7].
2. Biomarkers
Both EGFR and PTEN belong to the RTKs pathway, whose amplification or activation is the hallmark of primary GBM [17]. EGFR is a transmembrane receptor whose activation and dimerization triggers the phosphatidylinositol-3'-kinase (PIP3)-Akt-mammalian target of rapamycin (mTOR) cascade. This pathway plays a crucial role in cellular survival and apoptosis resistance [18]. PTEN is a tyrosine-phosphatase whose role is the inhibition of RTKs pathway [17]. EGFR amplification is the most common molecular alteration in primary GBM [16]. About two thirds of EGFR-amplified GBMs express EGFRvIII, a constitutively-activated, ligand-independent mutant form of EGFR [19]. PTEN mutation has been detected in 15-40% of primary GBM [17]. GBM is, by definition, a high vascular tumor [1] and therefore angiogenesis is a key mechanism for its maintenance and progression [20]. VEGF is the main regulator of angiogenesis in GBM; its production is triggered by tumor hypoxia and it is also released by GBM cancer stem cells in the “vascular niche” [20]. Moreover, VEGF exists in several isoforms with different biological properties [14].
3. Search Strategy
A search was performed in the PubMed database (http://www.ncbi.nlm.nih.gov/pubmed/) in February 2016, using the following key words, “glioblastoma”, “EGFRvIII”, “epidermal growth factor receptor variant III”, “VEGF”, “PTEN”, “mTOR”, “Akt”. Articles published after 2005, i.e. after radiochemotherapy with TMZ became the standard-of-care for GBM [3] were included in this review. Review articles as well as redundant publications were excluded from this review.
4. Results
4.1. Epidermal growth factor receptor variant III
Studies reporting the prognostic or predictive value of EGFRvIII are listed in Table 1[8,15,16,21-34]. We found 17 studies considering 1656 GBM patients overall. The main techniques for assessment of EGFRvIII expression were immunohistochemistry (12/17 studies, 70.6%) and PCR (8/17 studies, 47.1%). Two out of 17 (11.8%) studies (156 patients) reported a positive prognostic value of EGFRvIII expression. Three (17.6%) studies (740 patients) reported a negative prognostic value of EGFRvIII expression. Two (11.8%) studies (60 patients) reported a positive predictive value of EGFRvIII expression for response to TKIs. One (5.9%) study (49 patients) reported a negative predictive value of EGFRvIII expression for response to TKIs. Nine out of 17 (52.9%) studies (651 patients) reported no prognostic/predictive value of EGFRvIII expression.
Table 1. Studies evaluating prognostic and/or predictive role of EGFRvIII in GBM.
Author, year | No. GBM cases | Treatment | Assay Technique | Prognostic/predictive value | Proposed molecular mechanism |
---|---|---|---|---|---|
Heimberger et al, 2005 [21] | 196 | Stupp | IHC | ↓ (for long survivors) | cell proliferation, ependymal involvement |
Liu et al, 2005 [22] | 160 | Stupp | PCR | none | NA |
Heimberger et al, 2005 [23] | 54 | Stupp | IHC | none | NA |
Mellinghoff et al, 2005 [16] | 50 | erlotinib / gefitinib (recurrence) | IHC, PCR, western blotting | ↑ (erlotinib) | EGFRvIII/PTEN co-expression |
Pelloski et al, 2007 [24] | 509 | Stupp | IHC | ↓ (OS) | none |
Viana-Pereira et al, 2008 [25] | 27 | Stupp | IHC | none | NA |
Brown et al, 2008 [26] | 81 | Stupp + erlotinib | IHC | none | NA |
Van den Bent et al, 2009 [27] | 49 | erlotinib vs TMZ/BCNU | IHC | ↓ (erlotinib) | none |
Thiessen et al, 2009 [28] | 16 | lapatinib (recurrence) | PCR | none | NA |
Reardon et al, 2009 [29] | 20 | erlotinib + sirolimus (recurrence) | IHC | none | NA |
Uhm et al, 2011 [30] | 96 | RT+ gefitinib | IHC | none | NA |
Montano et al, 2011 [15] | 73 | Stupp | PCR | ↑ (OS) | increased effectiveness of TMZ in EGFRvIII-positive cells due to pathway addiction |
Lv et al, 2012 [31] | 35 | cetuximab | IHC, PCR | ↓ (OS, PFS) | none |
Bienkowski et al, 2013 [32] | 83 | RT or RT+CT | PCR, FISH | ↑ (OS) | none |
D’Alessandris et al, 2013 [8] | 10 | bev + erlotinib (recurrence) | PCR | ↑ (erlotinib) | tailored therapy |
Weller et al, 2014 [33] | 184 | Stupp | IHC, PCR, MLPA | none | possible false-negative testings |
Gallego et al, 2014 [34] | 13 recurrent | erlotinib | IHC | none | genetic heterogeneity of GBM |
negative prognostic/predictive value;
positive prognostic/predictive value.
bev, bevacizumab; BCNU, bis-chloroethylnitrosourea; CT, chemotherapy; EGFRvIII, epidermal growth factor receptor variant III; FISH, fluorescence in situ hybridization; IHC, immunohistochemistry; MLPA, multiplex ligation-dependent probe amplification; NA, not available; OS, overall survival; PCR, polymerase chain reaction; PTEN, phosphatase and tensin homolog; PFS, progression-free survival; RT, radiotherapy; TMZ, temozolomide.
4.2. Phosphatase and tensin homolog and other members of receptor tyrosine kinase pathway
Studies reporting the prognostic/predictive value of PTEN and of other members of RTK pathway are listed in Table 2 [15,16,26,28,29,35-45]. Overall, 16 studies enrolling 1927 patients could be found. PTEN expression was usually detected by immunohistochemistry (11/16 studies, 68.9%). PTEN expression had a positive prognostic value in 5/16 (31.3%) studies (285 patients), whereas no studies in which PTEN loss had a positive prognostic value were found. In 2/5 (40%) studies (98 patients) in which a positive prognostic role of PTEN expression could be demonstrated, iperactivation of EGFR pathway played a synergistic role. In one out of 16 (6.3%) studies (50 patients), PTEN expression had a positive predictive value for response to TKIs. In one other study (32 patients), a positive prognostic value of p-Akt expression was detected. Finally, in 9 out of 16 (56.3%) studies (1560 patients), no prognostic/predictive value of PTEN expression could be determined.
Table 2. Studies evaluating prognostic and/or predictive role of PTEN/Akt/mTOR pathway in GBM.
Author, year | No. GBM cases | Treatment | Assay Technique | Prognostic/predictive value | Proposed molecular mechanism |
---|---|---|---|---|---|
Mellinghoff et al, 2005 [16] | 49 | Erlotinib/gefitinib (recurrence) | PCR, IHC | PTEN: ↑ (for erlotinib) | EGFRvIII/PTEN co-expression |
Ohgaki and Kleihues, 2005 [35] | 680 | Stupp | NA | PTEN: none | NA |
Rich et al, 2005 [36] | 41 | Stupp | PCR, microarray | PTEN: none | NA |
Liu et al, 2006 [37] | 25 | NA | IHC | PTEN loss+ EGFR amplification: ↓ (OS) | none |
Homma et al, 2006 [38] | 420 | Stupp | PCR | PTEN: none | NA |
Fukushima et al, 2006 [39] | 63 | Stupp | SSCP, sequencing | PTEN: none | NA |
De Groot et al, 2008 [40] | 43 | Carboplatin + erlotinib (for recurrence) | IHC | PTEN: none | NA |
Brown et al, 2008 [26] | 81 | Stupp +Erlotinib | IHC | PTEN: none | NA |
Thiessen et al, 2010 [28] | 16 | Lapatinib (recurrence) | IHC | PTEN: none | NA |
Umesh et al, 2009 [41] | 54 | Stupp | IHC | PTEN loss: ↓ (OS) | associated EGFR expression |
Ruano et al, 2009 [42] | 194 | Stupp | IHC | PTEN: none | NA |
Kreisl et al, 2009 [43] | 22 | Gefitinib + everolimus (recurrence) | IHC | PTEN: none | NA |
Reardon et al, 2010 [29] | 32 | Erlotinib+ Sirolimus (recurrence) | IHC | p-AKT +: ↑ (PFS) | unclear. Possible laboratory errors |
Montano et al, 2011 [15] | 73 | Stupp | IHC | PTEN normal + EGFRvIII positive: ↑ (OS) | tumor suppression role of PTEN |
Srividya et al, 2013 [44] | 73 | Stupp | FISH for homozygous deletion of 10q23/PTEN | PTEN loss: ↓ (OS) | Tumor suppression role of PTEN |
Idoate et al, 2014 [45] | 60 | Stupp | IHC, PCR | PTEN loss: ↓ (OS) | Tumor suppression role of PTEN |
negative prognostic/predictive value;
positive prognostic/predictive value.
EGFR, epidermal growth factor receptor; EGFRvIII, EGFR variant III; FISH, fluorescence in situ hybridization; IHC, immunohistochemistry; NA, not available; OS, overall survival; PCR, polymerase chain reaction; PFS, progression-free survival; PTEN, phosphatase and tensin homolog; SSCP, single-strand conformation polymorphism.
4.3. Vascular endothelial growth factor
Details on 11 studies (445 patients) analyzing the prognostic/predictive value of VEGF expression are given in Table 3 [8,14,46-54]. VEGF expression was mostly analyzed by immunohistochemistry (7/11 studies, 63.6%). Overexpression of VEGF had a positive prognostic value in one out of 11 (9.1%) studies (27 patients) and a negative prognostic value in 3/11 (27.3) studies (183 patients). Overexpression of VEGF was predictive of response to anti-angiogenic therapy in 2/11 (18.2%) studies (37 patients). Circulating VEGF levels carried a negative prognostic value in one study (47 patients). One study on 25 GBM patients assessed the predictive value of tumor VEGF isoforms for response to anti-angiogenic therapy. Finally, in 3/17 (27.3%) studies (126 patients) VEGF expression had no prognostic/predictive value.
Table 3. Studies evaluating prognostic and/or predictive role of VEGF in GBM.
Author, year | No. GBM cases | Treatment | Assay Technique | Prognostic/predictive value | Proposed molecular mechanism |
---|---|---|---|---|---|
Tuettenberg et al, 2005 [46] | 12 | Stupp + rofecoxib | IHC | none | NA |
Pope et al, 2008 [47] | 52 | Stupp | micro-array | none | NA |
Flynn et al, 2008 [48] | 62 | Stupp | IHC | Overexpression: ↓ (for OS) | GLUT-1 coexpression, hypoxia |
Kesari et al, 2008 [49] | 47 | Stupp + thalidomide and celecoxib | ELISA | High serum levels: ↓ (for OS) | none |
Sathornsumetee et al, 2008 [50] | 27 | bev + CPT-11 (recurrence) | IHC | Overexpression: ↑ (for response) | targeted therapy |
Reardon et al, 2009 [51] | 27 | bev + etoposide (recurrence) | IHC | Overexpression: ↑ (for OS) | targeted therapy |
Sie et al, 2009 [52] | 62 | Stupp | IHC | none | NA |
D’Alessandris et al, 2013 [8] | 10 | bev + erlotinib (recurrence) | IHC | overexpression: ↑ (for response) | tailored therapy |
D’Alessandris et al, 2015 [14] | 25 | bev (recurrence) | PCR | total VEGF and VEGF-121: ↓ (for PFS) | Heavier VEGF isoforms are the main target of bev |
Irshad et al, 2015 [53] | 35 | NA | PCR | activation of hypoxia cascade (incl. VEGF): ↓ (for OS) | Oncogenic role of hypoxia |
Zhao et al 2016 [54] | 86 | Stupp | IHC | VEGF-C overexpression: ↓ (for OS) | VEGF-C stimulates NRP2 in paracrine/ autocrine loop |
negative prognostic/predictive value;
positive prognostic/predictive value.
bev, bevacizumab; CPT-11, irinotecan; ELISA, enzyme linked immunosorbent assay; IHC, immunohistochemistry; NA, not available; NRP2, neuropilin-2;
OS, overall survival; PCR, polymerase chain reaction; PFS, progression-free survival; RT, radiotherapy; TMZ, temozolomide; VEGF, vascular endothelial growth factor.
5. Discussion
The aim of this review was to perform a thorough, up-to-date review of prognostic and predictive value of EGFRvIII expression, PTEN deletion and other alteration of the RTK pathway, and VEGF expression. We decided to focus our work on these markers because of their importance in gliomagenesis, their role as potential targets for individualized therapies and because of their translational relevance is currently under debate. EGFRvIII is a constitutively activated mutant form of EGFR. Traditionally, EGFRvIII has been considered an oncogenic protein [19], due to preclinical studies in which EGFRvIII-expressing cells had a superior invasiveness, proliferation and tumorigenesis potential [55,56]. The majority of studies listed in Table 1 are in accordance with this assumption. Nonetheless, in these studies EGFRvIII expression was studied mainly by immunohistochemistry, whose reliability as assay method has been showed inferior to PCR [57]. Interestingly, the studies which demonstrated a positive prognostic and a predictive role for response to TKIs of EGFRvIII adopted the semiquantitative PCR determination as assay method. This discrepancy could be explained by the fact that very low levels of EGFRvIII can be detected by PCR as opposed to immunohistochemistry where the specimens would likely be determined as negative. As concerns PTEN, there is a general agreement on the positive prognostic role of its retained expression. Of note, a tight interaction of PTEN and EGFR expression is documented in several studies. Taken cumulatively, studies listed in Table 1 and Table 2 appear in line with the pioneering study by Mellinghoff et al. [16], in which co-expression of EGFRvIII and PTEN had a positive predictive value to response to TKIs. However, this hypothesis probably deserves further validation, at least in the setting of recurrent GBM [15]. Overexpression of VEGF appears to carry a negative prognostic value in term of survival but a positive predictive value to response to anti-angiogenic therapy. Unfortunately, this evidence has been insufficiently tested in the large phase III trials with bevacizumab, conducted on newly diagnosed GBM [5,6]. Interestingly, Kesari et al. [49] evaluated the prognostic role of serum VEGF levels. This kind of assay is widely used in other tumors [58], but evidence regarding GBM is low. Another promising research field, which has been pioneered by our group, is the analysis of the role of VEGF isoforms [14], which originate from alterative splicing of the VEGF gene and have different biological properties [59]. In detail, the heavier isoforms (VEGF-206, VEGF-189) are bound to extracellular matrix and act both as VEGF reserve and as trigger of local angiogenesis. The lighter VEGF-121 is freely diffusible but has a weaker biological activity; the intermediate-weight VEGF-165 has also intermediate properties. Consistently with these premises, it has been showed that patients with high level of VEGF-121 responded poorly to bevacizumab, probably because in these patients a lesser residual amount of bevacizumab is available to target the heavier and more active VEGF isoforms [14].
Taken altogether, the results of our study show that none of the markers analyzed in this review reached sufficient evidence to be considered a clear prognosticator for survival. Moreover none of these potential biomarkers was clearly predictive of response to adjuvant therapy. This data is particularly unpleasant in the setting of recurrent GBM, in which there is a strong need of predictive biomarkers for response to targeted therapy [7]. The number of studies in which no role for biomarkers could be detected is also troublesome; this can reflect both GBM heterogeneity and the inaccuracy of the assay methods employed.
In order to improve biomarkers detection and validation, current and future clinical trials need to prospectively assess their potential role. Several subgroups should be designed, with the aim of tailoring the treatment on patient’s molecular profile. This claims for large, collaborative, multicenter trials, able to reach adequate recruitment targets. In order to minimize biomarkers determination errors, a few pathology “reference” laboratories for each country should be identified that intratumor heterogeneity can be solved by analyzing several tumor samples from different regions. When applicable, analysis of cancer stem cells profile can provide a reliable picture of the tumor’s landscape [60]. A new and intriguing opportunity for studying the genomic and/or proteomic profile of GBM for prognostic or predictive purposes is provided by the so-called “liquid biopsies”, i.e. the analysis of peripheral blood samples. The main target of these biopsies are the circulating tumor cells, which can be analyzed using standard immunocytochemical or molecular biology techniques. Liquid biopsies are currently used for prognostic/predictive purposes in several tumors. In GBM, the difficulty to identify affordable biomarkers to separate circulating tumor cells from normal blood cells [61] has hindered the development of this technique. Alternatives to circulating tumor cells, to be analyzed in a liquid biopsy, include RNA sequencing of “tumor-educated platelets” [62] and genomic profiling of microvescicles [63].
Understanding the gliomagenesis pathways and looking for biomarkers endowed with translational relevance are hard tasks in a heterogeneous tumor like GBM. However, this is a necessary effort in order to find the appropriate, tailored therapy for each specific GBM patient.
Disclosures
The authors reported no conflict of interest.
References
- [1].Louis DN, Ohgaki H, Cavenee WK, editors. International Agency for Research on Cancer; Lyon: 2007. WHO classification of Tumours of the Central Nervous System. [Google Scholar]
- [2].Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro Oncol. 2012;14(Suppl 5):v1–v49. doi: 10.1093/neuonc/nos218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO. European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
- [4].Weller M, van den Bent M, Hopkins K, Tonn JC, Stupp R, Falini A, Cohen-Jonathan-Moyal E, Frappaz D, Henriksson R, Balana C, Chinot O, Ram Z, Reifenberger G, Soffietti R, Wick W. European Association for Neuro-Oncology (EANO) Task Force on Malignant Glioma. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol. 2014;15(9):e395–403. doi: 10.1016/S1470-2045(14)70011-7. [DOI] [PubMed] [Google Scholar]
- [5].Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, Brandes AA, Hilton M, Abrey L, Cloughesy T. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709–722. doi: 10.1056/NEJMoa1308345. [DOI] [PubMed] [Google Scholar]
- [6].Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, Jeraj R, Brown PD, Jaeckle KA, Schiff D, Stieber VW, Brachman DG, Werner-Wasik M, Tremont-Lukats IW, Sulman EP, Aldape KD, Curran WJ, Jr, Mehta MP. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699–708. doi: 10.1056/NEJMoa1308573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Weller M, Yung WK. Angiogenesis inhibition for glioblastoma at the edge: beyond AVAGlio and RTOG 0825. Neuro Oncol. 2013;15:971. doi: 10.1093/neuonc/not106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].D'Alessandris QG, Montano N, Cenci T, Martini M, Lauretti L, Bianchi F, Larocca LM, Maira G, Fernandez E, Pallini R. Targeted therapy with bevacizumab and erlotinib tailored to the molecular profile of patients with recurrent glioblastoma. Preliminary experience. Acta Neurochir (Wien) 2013;155:33–40. doi: 10.1007/s00701-012-1536-5. [DOI] [PubMed] [Google Scholar]
- [9].Louis DN, Perry A, Burger P, Ellison DW, Reifenberger G, von Deimling A, Aldape K, Brat D, Collins VP, Eberhart C, Figarella-Branger D, Fuller GN, Giangaspero F, Giannini C, Hawkins C, Kleihues P, Korshunov A, Kros JM, Beatriz Lopes M, Ng HK, Ohgaki H, Paulus W, Pietsch T, Rosenblum M, Rushing E, Soylemezoglu F, Wiestler O, Wesseling P. International Society Of Neuropathology--Haarlem. International Society of Neuropathology—Haarlem consensus guidelines for nervous system tumor classification and grading. Brain Pathol. 2014;24:429–435. doi: 10.1111/bpa.12171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JEC, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997–1003. doi: 10.1056/NEJMoa043331. [DOI] [PubMed] [Google Scholar]
- [11].D’Alessandris QG, Montano N, Larocca LM, Maira G, Pallini R. Prognostic Impact of MGMT Promoter Methylation in Glioblastoma – A Systematic Review. J Cancer Sci Ther. 2014;6:136–141. [Google Scholar]
- [12].Ohgaki H, Kleihues P. The Definition of Primary and Secondary Glioblastoma. Clin Cancer Res. 2013;19:764–772. doi: 10.1158/1078-0432.CCR-12-3002. [DOI] [PubMed] [Google Scholar]
- [13].Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344:1396–1401. doi: 10.1126/science.1254257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].D'Alessandris QG, Martini M, Cenci T, Capo G, Ricci-Vitiani L, Larocca LM, Pallini R. VEGF isoforms as outcome biomarker for anti-angiogenic therapy in recurrent glioblastoma. Neurology. 2015;84:1906–1908. doi: 10.1212/WNL.0000000000001543. [DOI] [PubMed] [Google Scholar]
- [15].Montano N, Cenci T, Martini M, D'Alessandris QG, Pelacchi F, Ricci-Vitiani L, Maira G, De Maria R, Larocca LM, Pallini R. Expression of EGFRvIII in glioblastoma: prognostic significance revisited. Neoplasia. 2011;13:1113–1121. doi: 10.1593/neo.111338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ, Lu KV, Yoshimoto K, Huang JH, Chute DJ, Riggs BL, Horvath S, Liau LM, Cavenee WK, Rao PN, Beroukhim R, Peck TC, Lee JC, Sellers WR, Stokoe D, Prados M, Cloughesy TF, Sawyers CL, Mischel PS. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005;353:2012–2024. doi: 10.1056/NEJMoa051918. [DOI] [PubMed] [Google Scholar]
- [17].Ohgaki H, Kleihues P. Genetic Pathways to Primary and Secondary Glioblastoma. Am J Pathol. 2007;170:1445–1453. doi: 10.2353/ajpath.2007.070011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Van den Bent MJ, Kros JM. Predictive and Prognostic Markers in Neuro-Oncology. J Neuropathol Exp Neurol. 2007;66:1074–1081. doi: 10.1097/nen.0b013e31815c39f1. [DOI] [PubMed] [Google Scholar]
- [19].Gan HK, Kaye AH, Luwor RB. The EGFRvIII variant in glioblastoma multiforme. J Clin Neurosci. 2009;16:748–754. doi: 10.1016/j.jocn.2008.12.005. [DOI] [PubMed] [Google Scholar]
- [20].Ricci-Vitiani L, Pallini R, Biffoni M, Todaro M, Invernici G, Cenci T, Maira G, Parati EA, Stassi G, Larocca LM, De Maria R. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature. 2010;468:824–828. doi: 10.1038/nature09557. [DOI] [PubMed] [Google Scholar]
- [21].Heimberger AB, Hlatky R, Suki D, Yang D, Weinberg J, Gilbert M, Sawaya R, Aldape K. Prognostic effect of epidermal growth factor receptor and EGFRvIII in glioblastoma multiforme patients. Clin Cancer Res. 2005;11:1462–1466. doi: 10.1158/1078-0432.CCR-04-1737. [DOI] [PubMed] [Google Scholar]
- [22].Liu L, Bäcklund LM, Nilsson BR, Grandér D, Ichimura K, Goike HM, Collins VP. Clinical significance of EGFR amplification and the aberrant EGFRvIII transcript in conventionally treated astrocytic gliomas. J Mol Med. 2005;83:917–926. doi: 10.1007/s00109-005-0700-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Heimberger AB, Suki D, Yang D, Shi W, Aldape K. The natural history of EGFR and EGFRvIII in glioblastoma patients. J Transl Med. 2005;3:38. doi: 10.1186/1479-5876-3-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Pelloski CE, Ballman KV, Furth AF, Zhang L, Lin E, Sulman EP, Bhat K, McDonald JM, Yung WK, Colman H, Woo SY, Heimberger AB, Suki D, Prados MD, Chang SM, Barker FG, 2nd, Buckner JC, James CD, Aldape K. Epidermal growth factor receptor variant III status defines clinically distinct subtypes of glioblastoma. J Clin Oncol. 2007;25:2288–2294. doi: 10.1200/JCO.2006.08.0705. [DOI] [PubMed] [Google Scholar]
- [25].Viana-Pereira M, Lopes JM, Little S, Milanezi F, Basto D, Pardal F, Jones C, Reis RM. Analysis of EGFR overexpression, EGFR gene amplification and the EGFRvIII mutation in Portuguese high-grade gliomas. Anticancer Res. 2008;28:913–920. [PubMed] [Google Scholar]
- [26].Brown PD, Krishnan S, Sarkaria JN, Wu W, Jaeckle KA, Uhm JH, Geoffroy FJ, Arusell R, Kitange G, Jenkins RB, Kugler JW, Morton RF, Rowland KM, Jr, Mischel P, Yong WH, Scheithauer BW, Schiff D, Giannini C, Buckner JC. North Central Cancer Treatment Group Study N0177. Phase I/II trial of erlotinib and temozolomide with radiation therapy in the treatment of newly diagnosed glioblastoma multiforme: North Central Cancer Treatment Group Study N0177. J Clin Oncol. 2008;26:5603–5609. doi: 10.1200/JCO.2008.18.0612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].van den Bent MJ, Brandes AA, Rampling R, Kouwenhoven MCM, Kros JM, Carpentier AF, Clement PM, Frenay M, Campone M, Baurain JF, Armand JP, Taphoorn MJB, Tosoni A, Kletzl H, Klughammer B. Lacombe D and Gorlia T. Randomized phase II trial of erlotinib versus temozolomide or car-Study. J Clin Oncol. 2009;27:579–584. doi: 10.1200/JCO.2008.17.5984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Thiessen B, Stewart C, Tsao M, Kamel-Reid S, Schaiquevich P, Mason W, Easaw J, Belanger K, Forsyth P, McIntosh L, Eisenhauer E. A phase I/II trial of GW572016 (lapatinib) in recurrent glioblastoma multiforme: clinical outcomes, pharmacokinetics and molecular correlation. Cancer Chemother Pharmacol. 2010;65:353–361. doi: 10.1007/s00280-009-1041-6. [DOI] [PubMed] [Google Scholar]
- [29].Reardon DA, Desjardins A, Vredenburgh JJ, Gururangan S, Friedman AH, Herndon JE, 2nd, Marcello J, Norfleet JA, McLendon RE, Sampson JH, Friedman HS. Phase 2 trial of erlotinib plus sirolimus in adults with recurrent glioblastoma. J Neurooncol. 2010;96:219–230. doi: 10.1007/s11060-009-9950-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Uhm JH, Ballman KV, Wu W, Giannini C, Krauss JC, Buckner JC, James CD, Scheithauer BW, Behrens RJ, Flynn PJ, Schaefer PL, Dakhill SR, Jaeckle KA. Phase II evaluation of gefitinib in patients with newly diagnosed Grade 4 astrocyto-ma: Mayo/North Central Cancer Treatment Group Study N0074. Int J Radiat Oncol Biol Phys. 2011;80:347–353. doi: 10.1016/j.ijrobp.2010.01.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Lv S, Teugels E, Sadones J, De Brakeleer S, Duerinck J, Du Four S, Michotte A, De Grève J, Neyns B. Correlation of EGFR, IDH1 and PTEN status with the outcome of patients with recurrent glioblastoma treated in a phase II clinical trial with the EGFR-blocking monoclonal antibody cetuximab. Int J Oncol. 2012;41:1029–1035. doi: 10.3892/ijo.2012.1539. [DOI] [PubMed] [Google Scholar]
- [32].Bieńkowski M, Piaskowski S, Stoczyńska-Fidelus E, Szybka M, Banaszczyk M, Witusik-Perkowska M, Jesień-Lewandowicz E, Jaskólski DJ, Radomiak-Załuska A, Jesionek-Kupnicka D, Sikorska B, Papierz W, Rieske P, Liberski PP. Screening for EGFR amplifications with a novel method and their significance for the outcome of glioblastoma patients. PLoS One. 2013;8:e65444. doi: 10.1371/journal.pone.0065444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Weller M, Kaulich K, Hentschel B, Felsberg J, Gramatzki D, Pietsch T, Simon M, Westphal M, Schackert G, Tonn JC, von Deimling A, Davis T, Weiss WA, Loeffler M, Reifenberger G, German Glioma Network Assessment and prognostic signifi-cance of the epidermal growth factor receptor vIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy. Int J Cancer. 2014;134:2437–2447. doi: 10.1002/ijc.28576. [DOI] [PubMed] [Google Scholar]
- [34].Gallego O, Cuatrecasas M, Benavides M, Segura PP, Berrocal A, Erill N, Colomer A, Quintana MJ, Balaña C, Gil M, Gal-lardo A, Murata P. Barnadas A Efficacy of erlotinib in patients with relapsed gliobastoma multiforme who expressed EGFRVIII and PTEN determined by immunohistochemistry. J Neurooncol. 2014;116:413–419. doi: 10.1007/s11060-013-1316-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol. 2005;64:479–489. doi: 10.1093/jnen/64.6.479. [DOI] [PubMed] [Google Scholar]
- [36].Rich JN, Hans C, Jones B, Iversen ES, McLendon RE, Rasheed BK, Dobra A, Dressman HK, Bigner DD, Nevins JR, West M. Gene expression profiling and genetic markers in glioblastoma survival. Cancer Res. 2005;65:4051–4058. doi: 10.1158/0008-5472.CAN-04-3936. [DOI] [PubMed] [Google Scholar]
- [37].Liu JG, Liu YH, Cai J, Liu XS, Song WZ, Huang Y, Mao Q. Expression of epidermal growth factor receptor and PTEN in malignancy brain tumors. Sichuan Da Xue Xue Bao Yi Xue Ban. 2006;37:868–871. [PubMed] [Google Scholar]
- [38].Homma T, Fukushima T, Vaccarella S, Yonekawa Y, Di Patre PL, Franceschi S, Ohgaki H. Correlation among pathology, genotype, and patient outcomes in glioblastoma. J Neuropathol Exp Neurol. 2006;65:846–854. doi: 10.1097/01.jnen.0000235118.75182.94. [DOI] [PubMed] [Google Scholar]
- [39].Fukushima T, Favereaux A, Huang H, Shimizu T, Yonekawa Y, Nakazato Y, Ohagki H. Genetic alterations in primary glioblastomas in Japan. J Neuropathol Exp Neurol. 2006;65:12–8. doi: 10.1097/01.jnen.0000196132.66464.96. [DOI] [PubMed] [Google Scholar]
- [40].de Groot JF, Gilbert MR, Aldape K, Hess KR, Hanna TA, Ictech S, Groves MD, Conrad C, Colman H, Puduvalli VK, Levin V. Yung WK Phase II study of carboplatin and erlotinib (Tarceva, OSI-774) in patients with recurrent glioblastoma. J Neurooncol. 2008;90:89–97. doi: 10.1007/s11060-008-9637-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Umesh S, Tandon A, Santosh V, Anandh B, Sampath S, Chandramouli BA, Sastry Kolluri VR. Clinical and immunohistochemical prognostic factors in adult glioblastoma patients. Clin Neuropathol. 2009;28:362–372. [PubMed] [Google Scholar]
- [42].Ruano Y, Ribalta T, de Lope AR, Campos-Martín Y, Fiaño C, Pérez-Magán E, Hernández-Moneo JL, Mollejo M, Meléndez B. Worse outcome in primary glioblastoma multiforme with concurrent epidermal growth factor receptor and p53 alteration. Am J Clin Pathol. 2009;131:257–263. doi: 10.1309/AJCP64YBDVCTIRWV. [DOI] [PubMed] [Google Scholar]
- [43].Kreisl TN, Lassman AB, Mischel PS, Rosen N, Scher HI, Teruya-Feldstein J, Shaffer D, Lis E, Abrey LE. A pilot study of everolimus and gefitinib in the treatment of recurrent glioblastoma (GBM) J Neurooncol. 2009;92:99–105. doi: 10.1007/s11060-008-9741-z. [DOI] [PubMed] [Google Scholar]
- [44].Srividya MR, Thota B, Shailaja BC, Arivazhagan A, Thennarasu K, Chandramouli BA, Hegde AS, Santosh V. Homozygous 10q23/PTEN deletion and its impact on outcome in glioblastoma: a prospective translational study on a uniformly treated cohort of adult patients. Neuropathology. 2011;31:376–383. doi: 10.1111/j.1440-1789.2010.01178.x. [DOI] [PubMed] [Google Scholar]
- [45].Idoate MA, Echeveste J, Diez-Valle R, Lozano MD, Aristu J. Biological and clinical significance of the intratumour heterogeneity of PTEN protein expression and the corresponding molecular abnormalities of the PTEN gene in glioblastomas. Neuropathol Appl Neurobiol. 2014;40:736–746. doi: 10.1111/nan.12117. [DOI] [PubMed] [Google Scholar]
- [46].Tuettenberg J, Grobholz R, Korn T, Wenz F, Erber R, Vajkoczy P. Continuous low-dose chemotherapy plus inhibition of cyclooxygenase-2 as an antiangiogenic therapy of glioblastoma multiforme. J Cancer Res Clin Oncol. 2005;131:31–40. doi: 10.1007/s00432-004-0620-5. [DOI] [PubMed] [Google Scholar]
- [47].Pope WB, Chen JH, Dong J, Carlson MR, Perlina A, Cloughesy TF, Liau LM, Mischel PS, Nghiemphu P, Lai A, Nelson SF. Relationship between gene expression and enhancement in glioblastoma multiforme: exploratory DNA microarray analysis. Radiology. 2008;249:268–277. doi: 10.1148/radiol.2491072000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Flynn JR, Wang L, Gillespie DL, Stoddard GJ, Reid JK, Owens J, Ellsworth GB, Salzman KL, Kinney AY, Jensen RL. Hypoxia-regulated protein expression, patient characteristics, and preoperative imaging as predictors of survival in adults with glioblastoma multiforme. Cancer. 2008;113:1032–1042. doi: 10.1002/cncr.23678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Kesari S, Schiff D, Henson JW, Muzikansky A, Gigas DC, Doherty L, Batchelor TT, Longtine JA, Ligon KL, Weaver S, Laforme A, Ramakrishna N, Black PM, Drappatz J, Ciampa A, Folkman J, Kieran M, Wen PY. Phase II study of temozolomide, thalidomide, and celecoxib for newly diagnosed glioblastoma in adults. Neuro Oncol. 2008;10:300–308. doi: 10.1215/15228517-2008-005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Sathornsumetee S, Cao Y, Marcello JE, Herndon JE, 2nd, McLendon RE, Desjardins A, Friedman HS, Dewhirst MW, Vredenburgh JJ, Rich JN. Tumor angiogenic and hypoxic profiles predict radiographic response and survival in malignant astrocytoma patients treated with bevacizumab and irinotecan. J Clin Oncol. 2008;26:271–278. doi: 10.1200/JCO.2007.13.3652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Reardon DA, Desjardins A, Vredenburgh JJ, Gururangan S, Sampson JH, Sathornsumetee S, McLendon RE, Herndon JE, 2nd, Marcello JE, Norfleet J, Friedman AH, Bigner DD, Friedman HS. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: a phase II study. Br J Cancer. 2009;101:1986–1994. doi: 10.1038/sj.bjc.6605412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Sie M, Wagemakers M, Molema G, Mooij JJ, de Bont ES, den Dunnen WF. The angiopoietin 1/angiopoietin 2 balance as a prognostic marker in primary glioblastoma multiforme. J Neurosurg. 2009;110:147–155. doi: 10.3171/2008.6.17612. [DOI] [PubMed] [Google Scholar]
- [53].Irshad K, Mohapatra SK, Srivastava C, Garg H, Mishra S, Dikshit B, Sarkar C, Gupta D, Chandra PS, Chattopadhyay P, Sinha S, Chosdol K. A combined gene signature of hypoxia and notch pathway in human glioblastoma and its prognostic relevance. PLoS One. 2015;10:e0118201. doi: 10.1371/journal.pone.0118201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Zhao H, Hou C, Hou A, Zhu D. Concurrent Expression of VEGF-C and Neuropilin-2 Is Correlated with Poor Prognosis in Glioblastoma. Tohoku J Exp Med. 2016;238:85–91. doi: 10.1620/tjem.238.85. [DOI] [PubMed] [Google Scholar]
- [55].Nishikawa R, Ji XD, Harmon RC, Lazar CS, Gill GN, Cavenee WK, Huang HJS. A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci USA. 1994;91:7727–7731. doi: 10.1073/pnas.91.16.7727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Huang HS, Nagane M, Klingbeil CK, Lin H, Nishikawa R, Ji XD, Huang CM, Gill GN, Wiley HS, Cavenee WK. The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. J Biol Chem. 1997;272:2927–2935. doi: 10.1074/jbc.272.5.2927. [DOI] [PubMed] [Google Scholar]
- [57].Yoshimoto K, Dang J, Zhu S, Nathanson D, Huang T, Dumont R, Seligson DB, Yong WH, Xiong Z, Rao N, Winther H, Chakravarti A, Bigner DD, Mellinghoff IK, Horvath S, Cavenee WK, Cloughesy TF, Mischel PS. Development of a real-time RT-PCR assay for detecting EGFRvIII in glioblastoma samples. Clin Cancer Res. 2008;14:488–493. doi: 10.1158/1078-0432.CCR-07-1966. [DOI] [PubMed] [Google Scholar]
- [58].Lambrechts D, Lenz HJ, de Haas S, Carmeliet P, Scherer SJ. Markers of response for the antiangiogenic agent bevacizumab. J Clin Oncol. 2013;31:1219–1230. doi: 10.1200/JCO.2012.46.2762. [DOI] [PubMed] [Google Scholar]
- [59].Ferrara N. Binding to the Extracellular Matrix and Proteolytic Processing: Two Key Mechanisms Regulating Vascular Endothelial Growth Factor Action. Mol Biol Cell. 2010;21:687–690. doi: 10.1091/mbc.E09-07-0590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Pallini R, Ricci-Vitiani L, Banna GL, Signore M, Lombardi D, Todaro M, Stassi G, Martini M, Maira G, Larocca LM, De Maria R. Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin Cancer Res. 2008;14:8205–8212. doi: 10.1158/1078-0432.CCR-08-0644. [DOI] [PubMed] [Google Scholar]
- [61].Adamczyk LA, Williams H, Frankow A, Ellis HP, Haynes HR, Perks C, Holly JM, Kurian KM. Current Understanding of Circulating Tumor Cells-Potential Value in Malignancies of the Central Nervous System. Front Neurol. 2015;6:174. doi: 10.3389/fneur.2015.00174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, Schellen P, Verschueren H, Post E, Koster J, Ylstra B, Ameziane N, Dorsman J, Smit EF, Verheul HM, Noske DP, Reijneveld JC, Nilsson RJ, Tannous BA, Wesseling P, Wurdinger T. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell. 2015;28:666–676. doi: 10.1016/j.ccell.2015.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Westphal M, Lamszus K. Circulating biomarkers for gliomas. Nat Rev Neurol. 2015;11:556–566. doi: 10.1038/nrneurol.2015.171. [DOI] [PubMed] [Google Scholar]