Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 19.
Published in final edited form as: Cancer Invest. 2012 Jan;30(1):48–56. doi: 10.3109/07357907.2011.630050

Deregulated Signaling Pathways in Glioblastoma Multiforme: Molecular Mechanisms and Therapeutic Targets

Hua Mao 1, Drake G LeBrun 1, Jingxuan Yang 1, Vivian F Zhu 1, Min Li 1
PMCID: PMC3799884  NIHMSID: NIHMS511526  PMID: 22236189

Abstract

Glioblastoma multiforme (GBM) is the most malignant and aggressive type of brain tumor with an average life expectancy of less than 15 months. This is mostly due to the highly mutated genome of GBM, which is characterized by the deregulation of many key signaling pathways involving growth, proliferation, survival, and apoptosis. It is critical to explore novel diagnostic and therapeutic strategies that target these pathways to improve the treatment of malignant glioma in the future. This review summarizes the most common and important pathways that are highly mutated or deregulated in GBM and discusses potential therapeutic strategies targeting these pathways.

Keywords: Signal transduction, Therapy, GBM

INTRODUCTION

Gliomas are the most common type of primary brain tumor, and are histologically differentiated as astrocytomas, oligodendrogliomas, and ependymomas. The World Health Organization (WHO) classifies central nervous system tumors into four different grades: I, II, III and IV (1, 2). Grade IV glioblastoma multiforme (GBM) is the most frequent, devastating, and malignant astrocytic glioma. It is characterized by a high degree of cellularity, vascular proliferation, tumor cell chemoresistance, and necrosis (3, 4). Even after neurosurgical resection, followed by aggressive chemotherapy and radiotherapy, GBM is still considered an incurable malignancy (5). No effective treatment agent against GBM has been identified; thus, the median life expectancy of patients diagnosed with GBMs is about 14 months (3). Of all primary tumors, brain tumors constitute ∼2% and the annual global incidence rate of it is about seven per 100,000 individuals (6). In the USA, the overall incidence rate of primary brain tumors is 18.1 per 100,000 individuals. The overall prevalence rate was estimated to be 209.0 per 100,000 in 2004 and 221.8 per 100,000 in 2010 in the USA (7). Although the mechanisms of GBM onset and progression remain largely unknown, recent advances in the understanding of the signaling pathways that underlie GBM pathogenesis have led to the development of new therapeutic approaches targeting multiple oncogenic signaling aberrations associated with GBM. This review describes the signaling pathways that are physiologically and pathologically relevant to GBM and discusses some of the most promising therapeutic approaches attempting to target these pathways.

p53 SIGNALING PATHWAY IN GBM

The p53 pathway is the most commonly mutated pathway in tumorigenesis. A well-known tumor suppressor, p53 responds to DNA damage and various genotoxic and cytotoxic stresses by inducing cell cycle arrest and apoptosis. It not only plays a crucial role in tumor suppression, but is also a broad transcription factor. It regulates more than 2,500 genes (6, 8), most of which are involved in tumor development, tumorigenesis, and tumor invasion. GBMs are divided into primary and secondary subtypes. Primary GBMs develop quickly and robustly while secondary GBMs develop progressively from low-grade astrocytoma. p53 mutations play a particularly significant role in the development of secondary GBMs and are often the earliest detectable genetic alteration in primary brain tumors, as they are present in ∼65% of precursor low-grade diffuse astrocytomas. Mutations in the p53 pathway are also detected in primary gliomas, although less frequently (1, 9). Mutations in the p53 pathway occur through different mechanisms in primary versus secondary GBM. A majority of p53 mutations in secondary GBM have been shown to be located at two key codons 248 and 273; however, p53 mutations are more widely distributed in primary GBMs. These results suggest that p53 mutations in primary GBM arise as a secondary event due to general genomic instability of the GBM tumor microenvironment (9, 10).

MDM2 is an E3 ubiquitin ligase, which is also an important regulator of the p53 pathway. It negatively regulates p53 in two ways: transcriptional inhibition by direct binding, and degradation through its E3 ligase activity (1113). Amplification of MDM2 occurred in about 10% of all malignant GBMs, but only in tumors without a p53 mutation, indicating that MDM2 overexpression may provide alternative means for tumors to inactivate p53-regulated growth control without having to alter p53 itself (14). Similarly, MDM4 (MDMX), encoded by 1q32, is another important regulator of p53. Due to its overexpression in malignant gliomas (15, 16), MDM4 has been extensively studied to elucidate its potential therapeutic effects. Whether the amplification of MDM4 causes p53-dependent or independent human malignant glioma remains unclear, but one previous report suggested that overexpression of MDM4 could have possibly caused GBM via a different p53-independent growth control pathway (15), similar to that of MDM2.

The tumor suppressor protein ARF (p14ARF) is one of the upstream regulators of the p53 pathway. ARF regulates the transcriptional activities of p53 by directly binding to MDM2 and subsequently inhibiting its E3 ubiquitin ligase activity (17, 18). Inactivation or mutation of ARF was commonly found in both low-grade and high-grade gliomas (19). ARF and INK4a (p16INK4A), both encoded by the CDK2A locus, are crucial regulators of the key growth control pathways p53 and pRB, respectively. One of the most frequent mutations found in GBMs is the homozygous deletion of this locus (p16INK4a/p14ARF/p15INK4b) (20). Furthermore, in mouse model studies and analyses of human glioma samples, Labuhn et al. found that the co-deletion of ARF and INK4a increased accordingly with tumor progression from low- to high-grade gliomas (21). This finding suggests that the locus containing ARF and INK4a is a key contributor to GBM pathogenesis, and that deletion of this locus may be a fundamental event in the development of GBM (20).

Chromosomes 9p, 10q23.3, and 10q 25–26 are the most frequently mutated or deleted regions in human gliomas. These chromosomes encode several critical regulators of cell survival, growth, and apoptosis, including CDK2A, CDK2B, ARF, MDM2, EGFR, and PTEN. Overall, the p53 pathway is disrupted in approximately 80% of high-grade (WHO Grades III and IV) gliomas. Additionally, EGFR overexpression and PTEN mutations are also commonly found in high-grade gliomas (22, 23). As the most commonly mutated pathway in tumorigenesis, p53 represents a viable therapeutic target for the treatment of GBM. Increasing wild type p53 expression in tumor cells through the use of adenovirus-mediated p53 gene therapy is one possible treatment modality to overcome p53 mutation. In a phase I trial, Lang and colleagues (24) showed that intratumoral injection of a p53-containing adenovirus vector resulted in the transfer of the p53 gene and expression of functional exogenous p53 in all patients. However, transfected cells were only found within a short distance from the injection site, indicating no systemic viral dissemination. Two phase I trials evaluating the transduction efficiency and effectiveness of wild-type Ad5CMV-p53 gene therapy (NCT00004041) or recombinant adenovirus-p53 SCH-58500 (NCT00004080) in combination with surgery have also been completed (February 2009, data not published yet).

pRB SIGNALING PATHWAY IN GBM

The tumor suppressor retinoblastoma (pRB) plays a crucial role in inhibiting cell cycle progression by binding and inhibiting transcription factors of the E2F family. It is encoded by the RB gene, which is regulated by the complex of cyclin-dependent kinases (CDKs). The RB gene is located at 13q14.1-q14.2, which has been shown to be involved in the malignant progression of astrocytomas (25). Mutations in this area are detected in more than 20% of high-grade gliomas. The loss of pRB expression has been detected in GBM (26), and the loss of 13q was also associated with the transition from low- to intermediate-grade gliomas (25, 27). In the G1 phase, pRB is normally inactivated by Cyclin D/CDK4/CDK6-induced phosphorylation, which leads to the release of pRB from E2F and the subsequent stimulation of cell progression into the S phase. CDKN2B, which is a CDK inhibitor and is commonly inactivated in GBM, forms a complex with CDK4 or CDK6, thus preventing the activation of CDKs. The downstream effect of this inhibition is to prevent cell growth and the cell cycle through G1 progression by maintaining the activation state of pRB. In addition to the inactivation of CDKN2B, amplification of CDK4 and CDK6 in glioblastomas is also common, demonstrating that both CDK4 and CDK6 play pivotal roles in astrocytic tumorigenesis and glioma progression (28).

Since the pRB pathway is inactivated by the kinase activity of the CDK4/CDK6/Cyclin D complex, inhibition of CDK4/6 may be a novel chemotherapeutic treatment strategy in GBM patients with aberrantly expressed pRB. A phase II study (NCT01227434) is currently recruiting patients to determine the efficacy of PD 0332991, a novel small molecule inhibitor of CDK4 and CDK6, in patients with recurrent Rb-positive glioblastoma. Overall, the average pathway amplification rates in GBM are: EGFR 35.7%, GLI/CDK 4–13.4%, MDM 2–9.2%, and PIK3C2B/MDM 4–7.7%. The CDKN2A/ CDKN2B locus was deleted in 46.4% of the combined cases (16). Even though other groups have found different frequency rates for these gene mutations, the pRB and p53 pathways consistently appear to be among the most frequently mutated pathways in GBM. Therefore, treatment strategies targeting these signaling pathways may possess broad therapeutic potential in GBM.

PI3K–PTEN-Akt-mTOR Signaling Pathway In GBM

The PI3K-PTEN-Akt-mTOR pathway regulates normal cellular functions that can also be critical in tumorigenesis, including cellular proliferation, apoptosis, cell invasion, and mobility. Activation of phosphatidylinositol 3-kinase (PI3K) complex, the first intracellular member of this signaling pathway, is regulated by many growth factors in conjunction with their receptors, such as epidermal growth factor (EGF) and its receptor (EGFR). Activated PI3K generates phosphatidylinositol-3,4,5-triphosphate, which in turn activates Akt. Akt then activates mTOR, which integrates several upstream signals into effector actions on multiple downstream targets involved in cell growth and division (29). Tumor suppressors such as PTEN inhibit this pathway.

The PI3K complex links growth factors with cellular proliferation, differentiation, metabolism, and survival. Constitutive activation and mutation of the PI3K signaling pathway play crucial roles in the development of GBM (3032). The PI3K pathway is altered in about 70% of GBMs, either by deletion of PTEN or amplification of EGFR and/or vascular endothelial growth factor receptor (VEGFR)/platelet-derived growth factor receptor (PDGFR) alpha (33). Overexpression of EGFR is one of the most frequent signaling mutations in GBM, and its amplification leads to increased activation of the PI3K pathway (34, 35). EGFRvIII, an EGF ligand-independent mutant with the deletion of exons 2–7 of the EGFR gene, is another common mutation in GBM (36) and is a significant predictor of poor prognosis (37). The average amplification rate for EGFR in GBM is about 35% (16) and the rate for the EGFRvIII mutant is approximately 40% (6). Additionally, a close correlation exists between amplification of wild-type EGFR and over-expression of EGFRvIII mutant (37). Taken together, these data suggest that EGFR and EGFRvIII are viable targets for therapeutic approaches to GBMs (38).

mTOR (mammalian target of rapamycin) is composed of two parts, mTOR complexes 1 (mTORC1) and 2 (mTORC2), and acts both as a downstream effector and upstream regulator of PI3K, thus playing an important role in GBM tumorigenesis (32). mTORC1 promotes oncogenesis by driving the translation of oncogenes, inhibiting autophagy, upregulating hypoxia-inducible factor 1α to promote angiogenesis, and stimulating lipid uptake via activation of sterol regulatory element binding protein 1c (SREBP1c). Similarly, mTORC2 promotes cancer growth by stimulating glucose uptake via activation of Akt and activating serum/glucocorticoid regulated kinase (SGK), which assists in proliferation and survival (29). Previous reports demonstrated that activation of the mitogenic kinase pathway (MAPK) involves a feedback mechanism between mTOR and PI3K, supporting the argument for combined inhibitors in GBM treatment (39).

PTEN (phosphatase and tensin homologue, located on chromosome TEN) acts as a tumor suppressor gene, and mutations of PTEN lead to the development of many cancers including GBM. PTEN negatively regulates the PI3K/AKT/PKB pathway (40) by blocking Akt signaling via the reduction of intracellular levels of phosphatidylinositol-3,4,5-triphosphate. Thus, PTEN plays a critical role in regulating its downstream target genes including mTOR. Loss of heterozygosity of chromosome 10, which causes deletions or mutations of PTEN, is a common event in GBMs. It is likely that induced expression of functional PTEN may serve as a therapeutic strategy in GBMs with amplified or mutated EGFR-PI3K-Akt-mTOR pathways.

RAS/MAPK SIGNALING PATHWAY IN GBM

Human RAS genes (Rat Sarcoma) are transforming oncogenes, including three highly related genes called H-Ras, N-Ras, and K-Ras. RAS belongs to the G protein family; therefore, the activation and deactivation of RAS is controlled by its binding to guanosine triphosphate (GTP) or guanosine diphosphate (GDP), respectively (41, 42). Activated RAS further activates RAF kinase through direct binding, which then regulates downstream signaling pathways such as the mitogen-activated protein kinase (MAPK) pathway (43, 44). RAS also regulates the activities of other pathways including the PI3K pathway; consequently, RAS plays a pivotal role in regulating cell proliferation, differentiation, signal transduction, apoptosis, and tumorigenesis.

Growth factor receptors, such as EGFRs, PDGFRs, and receptor tyrosine kinases (RTKs) (45) have an additional role in regulating the activities of RAS through direct associations. Upon activation, RAS then activates RAF and phosphorylates a series of kinases including MEK and MAPK, through which RAS can regulate downstream target gene transcription and cell activities. Aberrant expression or defects in the RAS/MAPK pathway causes abnormal cell growth and proliferation, inducing other abnormal cellular activities such as invasion and apoptosis.

Many cancers have exhibited a defective RAS/MAPK pathway. In high-grade astrocytoma and GBM, the over-expression of RAS was detected in multiple cases (4648). Besides EGFR, other growth factor receptors that regulate RAS such as PDGFR or RTKs are also overexpressed, leading to the implication of a positive correlation between expression of PDGFR and RTKs and glioma pathogenesis. It is likely that the abnormal activation of the RAS/MAPK pathway could be a potential target for glioma treatment therapy.

STAT3 and ZIP4 SIGNALING IN GBM

STAT protein complexes (Signal transducers and activators of transcription) are a family of cytoplasmic proteins with SH2 (Src Homology-2) domains that act as transcription factors. STAT proteins regulate cellular responses to cytokines and growth factors by transducing signals from the plasma membrane to the nucleus (49), thus activating transcription of target genes and regulating cell activities such as proliferation, invasion, and apoptosis. STAT3 is one of the STAT family proteins that can be activated by EGF, and it is upregulated in multiple tumors including GBM (50). Past studies have indicated that STAT3 is a significant player in the development of neural stem cells and astrocytes (5153). Interestingly, STAT3 was once thought to possess only oncogenic properties, but it was recently found to function in opposing tumor suppressive or oncogenic roles in GBM, depending on the genetic profile of the tumor (54).

Zinc is an essential trace element and catalytic/structural component used by many metalloenzymes and zinc-dependent transcription factors. Recent studies indicate a possible correlation between zinc levels and cancer risk. Our previous studies have indicated that the zinc transporter, ZIP4, is overexpressed in human pancreatic cancer (55). Overexpressed ZIP4 also increases cell proliferation and zinc uptake of pancreatic cancer cells, indicating that aberrant ZIP4 upregulation may contribute to cancer pathogenesis and progression. While investigating the signal transduction pathway through which ZIP4 regulates cancer growth (56), our lab found that overexpression of ZIP4 increases IL-6 transcription through cAMP response element-binding (CREB), which in turn activates STAT3 and leads to increased cyclin D1 expression. This ultimately results in increased cell proliferation and tumor progression. Our recent data suggest that ZIP4 is overexpressed in GBM, and correlates with the overall survival GBM patients. Therefore, ZIP4 may modulate gliomagenesis and malignant gliomas by regulating the activity of STAT3 and its related molecules, making ZIP4 and STAT3 interesting therapeutic targets in controlling glioma malignancy and invasion.

TREATMENT OF GLIOBLASTOMA MULTIFORME

Despite a better understanding of the mechanisms that underlie the development of GBM gained in the last several decades, GBM treatment still lacks efficacy. The current standard of care for GBM includes surgical resection, followed by chemotherapy (mainly temozolomide (TMZ), a DNA alkylating agent) and/or radiation therapy. At present, the average survival rate for GBM is less than 15 months. Improving therapeutic efficacy remains a significant challenge in GBM treatment, and targeting the previously mentioned signaling pathways may enhance current GBM therapies and positively impact patient outcomes (Table 1).

Table 1.

Current clinical trials targeting key aberrant signaling pathways in GBM

Pathway Target Therapy Trial specifications Outcomes Reference
p53 p53 Ad-53 adenovirus Phase I (15 patients with recurrent
GBM)
Exogenous p53 detected within
tumor; no systemic
dissemination
Lang et al. 2003 (24)
p53 Ad5CMV-p53 Phase I (patients with recurrent
GBM)
Completed; not published
(NCT00004041)
p53 Ad-p53 SCH-58500 Phase I (patients with recurrent
GBM or AA)
Completed; not published
(NCT00004080)
pRB CDK4/6 PD 0332991 Phase II (patients with recurrent
pRB-positive GBM)
Recruiting (NCT01227434)
PI3K–mTOR-Akt-PTEN EGFR Erlotinib + RT + TMZ Phase II (65 patients with newly
diagnosed GBM vs. historical
control)
Median OS 19.3 vs. 14.1 months 1
(p = 0.01); Median PFS 8.2 vs.
4.9 months (p = 0.001)
Prados et al. 2009 (59)
EGFR Gefitinib Phase II (98 patients with newly
diagnosed GBM vs. historical
control)
OS-1 yr 54.2% vs. 48.9% (p > 0.05);
PFS-1 yr 16.7% vs. 30.3% (p >
0.05)
Uhm et al. 2011 (63)
EGFRvIII EGFRvIII vaccination Phase II (21 patients with
EGFRvIII-expressing newly
diagnosed GBM)
PFS-6 67%; Median PFS 14.2 vs. 6.3
months in control group (p = 0.041); Median OS 26.0 vs. 15.0
months (p = 0.001)
Heimberger et al. 2009 (91)
mTOR Temsirolimus Phase II (65 patients with recurrent
GBM)
Median PFS 2.3 months; Median
OS 4.4 months
Galanis et al. 2005 (68)
mTOR + PI3K XL765 + TMZ Phase I (patients with AA or GBM) Recruiting (NCT00704080)
VEGFR Bevacizumab Phase II (50 patients with recurrent
GBM)
PFS-6 25%; Median OS 25.6 weeks Raizer et al. 2010(78)
VEGFR Cediranib Phase II (31 patients with recurrent
GBM vs. historical control)
PFS-6 25.8% vs. 15%; Median PFS
117 vs. 63 days; Median OS 227
vs. 175 days
Batchelor et al. 2010(79)
VEGFR + PDGFR Sorafenib + TMZ Phase II (32 patients with recurrent
GBM)
PFS-6 9.4% Reardon et al. 2011 (82)
VEGFR + PDGFR Pazopanib Phase II (35 patients with recurrent
GBM)
Median PFS 12 weeks; Median OS
35 weeks
Iwamoto 2010 (92)
PDGFR Imatinib + hydroxyurea Phase II (231 patients with
recurrent GBM)
PFS-6 10.6%; Median OS 26.0
weeks
Reardon et al. 2009 (84)
Akt Enzastaurin Phase III (266 patients with
recurrent GBM)
PFS 1.5 vs. 1.6 months in lomustine
control; OS 6.6 vs. 7.1 months)
Wick et al. 2010(88)
Akt Nelfinavir + RT + TMZ Phase I Recruiting (NCT01020292)
Akt Perifosine Phase II Ongoing (NCT00590954)
RAS RAS Reovirus Phase I (12 patients with recurrent
GBM)
Median PFS 4.3 weeks; Median OS
21 weeks
Forsyth et al. 2008 (90)
RAS Reolysin Phase I/II Completed; not published
(NCT00528684)
RAS Zarnestra + TMZ Phase I/II Completed; not published
(NCT00050986)

The majority of clinical trials involving therapies targeting commonly deregulated signaling pathways in GBM have shown little to no clinical efficacy.

Abbreviations: AA, WHO Grade III anaplastic astrocytoma; GBM, WHO Grade IV glioblastoma multiforme; OS, Overall survival; OS-1 yr, Overall survival one year after initiation of treatment; PFS, Progression-free survival; PFS-6, Progression-free survival six months after initiation of treatment; RT, Radiation therapy; TMZ, Temozolomide.

TMZ

Temozolomide (TMZ), an oral alkylating and chemotherapeutic agent, was first used to treat primary brain tumors in 1993 (57) and became a major agent for treating primary brain tumors following surgical resection and radiotherapy. It alkylates or methylates DNA, causing cancer cells to die. O-6-methylguanine-DNA methyltransferase (MGMT) or O-6-alkylguanine-DNA alkyltransferase can diminish the effect of TMZ by repairing the DNA damage (58).

Previous studies have shown that GBMs are highly resistant to single inhibitor, suggesting that combinational strategies involving standard chemotherapies like TMZ and pathway inhibitors might be a possible future direction for treating GBM. Recently, TMZ has been combined with other inhibitors in many phase I and II clinical trials to try to improve the drug’s therapeutic efficacy. Temozolimide together with the EGFR inhibitor, erlotinib, and radiotherapy have recently been reported to improve patient survival time from 14.1 months to 19.3 months (59). The study showed PTEN expression correlates well with survival rates, suggesting that erlotinib regulates the PTEN/mTOR pathway. MGMT promotes methylation, which also correlates well with survival rates, suggesting a synergistic function between TMZ and erlotinib.

EGFR

As mentioned above, EGFRs are overexpressed in multiple GBM cases and play a significant role in regulating other intracellular signaling pathways that contribute to GBM pathogenesis including mTOR/PI3K/Akt and RAS/MAPK. Therefore, inhibiting EGFR may be a possible therapeutic strategy for GBM treatment. Many EGFR inhibitors, such as erlotinib, lapatinib, and gefitinib have been evaluated as single-agent therapies in phase I or phase II trials (6063), but these drugs had minimal activity in GBM patients and did not improve overall survival or progression-free survival. However, a phase II trial recently showed that vaccinating patients who possessed the EGFRvIII variant, one of the most frequent mutations in GBM, using an EGFRvIII-targeted peptide significantly prolonged progression-free survival (14.2 vs. 6.3 months) and overall survival (26 vs. 15 months) compared to a contemporaneously treated patient cohort matched for EGFRvIII expression and past treatment (64). A randomized, controlled phase III trial to thoroughly evaluate the vaccine is currently in the planning stages.

mTOR-PI3K-Akt-PTEN

Several mTOR-PI3K-Akt-PTEN signaling pathway inhibitors have been developed in the past decade. Among the most widely tested are mTOR inhibitors, including rapamycin and its intravenous and oral derivatives temosirolimus and everolimus, respectively. These drugs inhibit mTOR through the formation of a complex with FK-binding protein 12 (FKBP-12), which then binds to mTOR, preventing its activation and inhibiting tumor cell growth (65). Multiple studies have been conducted using each of these drugs, alone or in combination with other therapies, but results have been limited (6670). Numerous clinical trials are ongoing to further elucidate the effects of these mTOR inhibitors. Additionally, a phase I study (NCT00704080) is currently recruiting patients to evaluate the preliminary efficacy of XL765, a novel small molecule inhibitor of mTOR and PI3K, in combination with TMZ.

Vascular endothelial growth factor receptors (VEGFRs) and platelet-derived growth factor receptors (PDGFRs) contribute to the growth potential of tumors by activating the PI3K/Akt pathway (71, 72) and are possible targets for GBM treatment. Bevacizumab (Avastin), a vascular endothelial growth factor (VEGF) monoclonal antibody, was recently found to be effective for recurrent malignant glioma and inhibit angiogenesis by blocking VEGF-A (73). Aberrant vascular proliferation, necrosis, and infiltration of surrounding brain tissues are considered “hallmarks” of GBM. Blocking aberrant vascular proliferation by bevacizumab therapy could block the infiltration of high-grade gliomas. Recently, several groups reported the clinical trial results of bevacizumab alone or combined with different inhibitors such as carboplatin (a DNA alkylating reagent) (74) and erlotinib for treating GBM (7578). Bevacizumab reduced tumor size and prolonged progression-free survival (PFS) even though the overall survival rates remained unchanged.

Cediranib is another VEGFR inhibitor with anti-cancer potential. In a phase II study (79), cediranib monotherapy in patients with recurrent GBM resulted in a radiographic response proportion, progression-free survival, and overall survival that compared favorably with data from historical controls. A phase III, randomized trial (NCT00777153) is currently ongoing to assess the efficacy of cediranib in patients with recurrent GBM as a single-agent and in combination with lomustine, a DNA alkylating drug primarily used in brain tumor treatment. Other VEGFR/PDGFR inhibitors (80, 81) such as sorafenib (82), imatinib (83, 84), pazopanib (85), and vatalanib (86) have shown no significant clinical benefit due to their poor ability to penetrate the blood-brain barrier, which is an important cause of the high resistance of glioblastoma to chemotherapies. Several phase I and II trials are ongoing to further evaluate the efficacy of bevacizumab, cediranib, and other VEGFR/PDGFR inhibitors in the treatment of GBM.

Akt is another viable molecular target within the mTOR-PI3K-Akt-PTEN pathway, since high activity of Akt in GBM leads to increased cell survival and decreased apoptosis. Enzastaurin is a novel antineoplastic agent that acts on Akt by inhibiting protein kinase C-beta, which regulates Akt activation (87). However, enzastaurin was recently shown to possess no superior efficacy compared with lomustine in a phase III trial involving patients with recurrent GBM (88). Two recently developed Akt inhibitors, nelfinavir and perifosine, interfere with Akt by downregulating its phosphorylation (80, 81) and are being assessed in several ongoing phase I and II trials (NCT01020292, NCT00915694, NCT00590954).

RAS/MAPK

Constitutively activated RAS protein is one of the most common mutations involved in GBM tumorigenesis, and is a prime target for GBM chemotherapy. A recently completed phase I/II trial (NCT00050986) was conducted to evaluate the safety and efficacy of Zarnestra (tipifarnib), a farnesylation inhibitor that interferes with the post-translational processing of RAS, in combination with TMZ. Oncolytic viral vectors also target RAS mutation by selectively replicating in and killing cancer cells with an activated Ras pathway (89). In a phase I trial (90), Forsyth and colleagues showed that intratumoral administration of reovirus, an oncolytic virus, in patients with recurrent malignant gliomas led to a median survival of 21 weeks (range 6–234) and median time-to-progression of 4.3 weeks (range 2.6–39). Another phase I/II study (NCT00528684) evaluating an engineered reovirus (Reolysin, Oncolytics Biotech) in patients with malignant glioma was recently completed, but results have not been published.

CONCLUSION

The combination of inhibitors of multiple pathways and other therapies is considered one of many future directions for GBM treatment. In this review, we have described the major pathways that are highly mutated or deregulated in GBM as well as some newly developed therapies targeting those pathways. With the knowledge that we have learned from the human tumor biology, we believe that generating GBM mouse models, which mimic human GBM, could provide insight into potentially active drug combinations for future treatment. Inhibitors plus shRNA or small molecules that block/activate major pathways can be tested by using these GBM models, which will help us to elucidate the mechanisms that underlie the tumorigenesis and progression of GBM and eventually discover effective therapies for treating GBM.

ACKNOWLEDGMENTS

This work was supported in part by the National Institutes of Health (NIH) grant R21CA133604, R01CA138701, the William and Ella Owens Medical Research Foundation (M. Li), and the Vivian L. Smith Department of Neurosurgery at the University of Texas Health Science Center at Houston, Medical School.

Footnotes

DECLARATION OF INTEREST

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

REFERENCES

  • 1.St Louis DC, Woodcock JB, Fransozo G, Blair PJ, Carlson LM, Murillo M, Wells MR, Williams AJ, Smoot DS, Kaushal S, Grimes JL, Harlan D, Chute JP, June CH, Siebenlist U, Lee KP. Evidence for distinct intracellular signaling pathways in CD34+ progenitor to dendritic cell differentiation from a human cell line model. J Immunol. 1999;162:3237–3248. [PubMed] [Google Scholar]
  • 2.Kleihues P, Burger PC, Scheithauer BW. The new WHO classification of brain tumours. Brain Pathol. 1993;3:255–268. doi: 10.1111/j.1750-3639.1993.tb00752.x. [DOI] [PubMed] [Google Scholar]
  • 3.Ohgaki H, Kleihues P. Epidemiology and etiology of gliomas. Acta Neuropathol. 2005;109:93–108. doi: 10.1007/s00401-005-0991-y. [DOI] [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;64:479–489. doi: 10.1093/jnen/64.6.479. [DOI] [PubMed] [Google Scholar]
  • 5.Denysenko T, Gennero L, Roos MA, Melcarne A, Juenemann C, Faccani G, Morra I, Cavallo G, Reguzzi S, Pescarmona G, Ponzetto A. Glioblastoma cancer stem cells: heterogeneity, microenvironment and related therapeutic strategies. Cell Biochem Funct. 2010;28:343–351. doi: 10.1002/cbf.1666. [DOI] [PubMed] [Google Scholar]
  • 6.Huang PH, Cavenee WK, Furnari FB, White FM. Uncovering therapeutic targets for glioblastoma: a systems biology approach. Cell Cycle. 2007;6:2750–2754. doi: 10.4161/cc.6.22.4922. [DOI] [PubMed] [Google Scholar]
  • 7.Porter KR, McCarthy BJ, Freels S, Kim Y, Davis FG. Prevalence estimates for primary brain tumors in the United States by age, gender, behavior, and histology. Neuro Oncol. 2010;12:520–527. doi: 10.1093/neuonc/nop066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Huse JT, Holland EC. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat Rev Cancer. 2010;10:319–331. doi: 10.1038/nrc2818. [DOI] [PubMed] [Google Scholar]
  • 9.Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C, Schüler D, Probst-Hensch NM, Maiorka PC, Baeza N, Pisani P, Yonekawa Y, Yasargil MG, Lütolf UM, Kleihues P. Genetic pathways to glioblastoma: a population-based study. Cancer Res. 2004;64:6892–6899. doi: 10.1158/0008-5472.CAN-04-1337. [DOI] [PubMed] [Google Scholar]
  • 10.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]
  • 11.Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387:296–299. doi: 10.1038/387296a0. [DOI] [PubMed] [Google Scholar]
  • 12.Kubbutat MH, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature. 1997;387:299–303. doi: 10.1038/387299a0. [DOI] [PubMed] [Google Scholar]
  • 13.Itahana K, Mao H, Jin A, Itahana Y, Clegg HV, Lindström MS, Bhat KP, Godfrey VL, Evan GI, Zhang Y. Targeted inactivation of Mdm2 RING finger E3 ubiquitin ligase activity in the mouse reveals mechanistic insights into p53 regulation. Cancer Cell. 2007;12:355–366. doi: 10.1016/j.ccr.2007.09.007. [DOI] [PubMed] [Google Scholar]
  • 14.Reifenberger G, Ichimura K, Reifenberger J, Elkahloun AG, Meltzer PS, Collins VP. Refined mapping of 12q13-q15 amplicons in human malignant gliomas suggests CDK4/SAS and MDM2 as independent amplification targets. Cancer Res. 1996;56:5141–5145. [PubMed] [Google Scholar]
  • 15.Riemenschneider MJ, Büschges R, Wolter M, Reifenberger J, Boström J, Kraus JA, Schlegel U, Reifenberger G. Amplification and overexpression of the MDM4 (MDMX) gene from 1q32 in a subset of malignant gliomas without TP53 mutation or MDM2 amplification. Cancer Res. 1999;59:6091–6096. [PubMed] [Google Scholar]
  • 16.Rao SK, Edwards J, Joshi AD, Siu IM, Riggins GJ. A survey of glioblastoma genomic amplifications and deletions. J Neurooncol. 2010;96:169–179. doi: 10.1007/s11060-009-9959-4. [DOI] [PubMed] [Google Scholar]
  • 17.Kamijo T, Weber JD, Zambetti G, Zindy F, Roussel MF, Sherr CJ. Functional and physical interactions of the ARF tumor suppressor with p53 and Mdm2. Proc Natl Acad Sci USA. 1998;95:8292–8297. doi: 10.1073/pnas.95.14.8292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell. 1998;92:725–734. doi: 10.1016/s0092-8674(00)81401-4. [DOI] [PubMed] [Google Scholar]
  • 19.Nakamura M, Watanabe T, Klangby U, Asker C, Wiman K, Yonekawa Y, Kleihues P, Ohgaki H. p14ARF deletion and methylation in genetic pathways to glioblastomas. Brain Pathol. 2001;11:159–168. doi: 10.1111/j.1750-3639.2001.tb00388.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Solomon DA, Kim JS, Jean W, Waldman T. Conspirators in a capital crime: co-deletion of p18INK4c and p16INK4a/p14ARF/p15INK4b in glioblastoma multiforme. Cancer Res. 2008;68:8657–8660. doi: 10.1158/0008-5472.CAN-08-2084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Labuhn M, Jones G, Speel EJM, Maier D, Zweifel C, Gratzl O, Van Meir EG, Hegi ME, Merlo A. Quantitative real-time PCR does not show selective targeting of p14(ARF) but concomitant inactivation of both p16(INK4A) and p14(ARF) in 105 human primary gliomas. Oncogene. 2001;20:1103–1109. doi: 10.1038/sj.onc.1204197. [DOI] [PubMed] [Google Scholar]
  • 22.Lespagnard L, Gancberg D, Rouas G, Leclercq G, de Saint-Aubain Somerhausen N, Di Leo A, Piccart M, Verhest A, Larsimont D. Tumor-infiltrating dendritic cells in adenocarcinomas of the breast: a study of 143 neoplasms with a correlation to usual prognostic factors and to clinical outcome. Int J Cancer. 1999;84:309–314. doi: 10.1002/(sici)1097-0215(19990621)84:3<309::aid-ijc19>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 23.Fulci G, Labuhn M, Maier D, Lachat Y, Hausmann O, Hegi ME, Janzer RC, Merlo A, Van Meir EG. p53 gene mutation and ink4a-arf deletion appear to be two mutually exclusive events in human glioblastoma. Oncogene. 2000;19:3816–3822. doi: 10.1038/sj.onc.1203700. [DOI] [PubMed] [Google Scholar]
  • 24.Lang FF, Bruner JM, Fuller GN, Aldape K, Prados MD, Chang S, Berger MS, McDermott MW, Kunwar SM, Junck LR, Chandler W, Zwiebel JA, Kaplan RS, Yung WK. Phase I trial of adenovirus-mediated p53 gene therapy for recurrent glioma: biological and clinical results. J Clin Oncol. 2003;21:2508–2518. doi: 10.1200/JCO.2003.21.13.2508. [DOI] [PubMed] [Google Scholar]
  • 25.Henson JW, Schnitker BL, Correa KM, von Deimling A, Fassbender F, Xu HJ, Benedict WF, Yandell DW, Louis DN. The retinoblastoma gene is involved in malignant progression of astrocytomas. Ann Neurol. 1994;36:714–721. doi: 10.1002/ana.410360505. [DOI] [PubMed] [Google Scholar]
  • 26.Tortosa A, Ino Y, Odell N, Swilley S, Sasaki H, Louis DN, Henson JW. Molecular genetics of radiographically defined de novo glioblastoma multiforme. Neuropathol Appl Neurobiol. 2000;26:544–552. doi: 10.1046/j.0305-1846.2000.00290.x. [DOI] [PubMed] [Google Scholar]
  • 27.Bahuau M, Vidaud D, Jenkins RB, Bièche I, Kimmel DW, Assouline B, Smith JS, Alderete B, Cayuela JM, Harpey JP, Caille B, Vidaud M. Germ-line deletion involving the INK4 locus in familial proneness to melanoma and nervous system tumors. Cancer Res. 1998;58:2298–2303. [PubMed] [Google Scholar]
  • 28.Lam PY, Di Tomaso E, Ng HK, Pang JC, Roussel MF, Hjelm NM. Expression of p19INK4d, CDK4, CDK6 in glioblastoma multiforme. Br J Neurosurg. 2000;14:28–32. doi: 10.1080/02688690042870. [DOI] [PubMed] [Google Scholar]
  • 29.Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12:21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cancer Genome Atlas Research Network, 231 Collaborators. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA, Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321:1807–1812. doi: 10.1126/science.1164382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Akhavan D, Cloughesy TF, Mischel PS. mTOR signaling in glioblastoma: lessons learned from bench to bedside. Neuro Oncol. 2010;12:882–889. doi: 10.1093/neuonc/noq052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhi YH, Song MM, Wang PL, Zhang T, Yin ZY. Suppression of matrix metalloproteinase-2 via RNA interference inhibits pancreatic carcinoma cell invasiveness and adhesion. World J Gastroenterol. 2009;15:1072–1078. doi: 10.3748/wjg.15.1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Peraud A, Watanabe K, Plate KH, Yonekawa Y, Kleihues P, Ohgaki H. p53 mutations versus EGF receptor expression in giant cell glioblastomas. J Neuropathol Exp Neurol. 1997;56:1236–1241. doi: 10.1097/00005072-199711000-00008. [DOI] [PubMed] [Google Scholar]
  • 35.Watanabe K, Tachibana O, Sata K, Yonekawa Y, Kleihues P, Ohgaki H. Overexpression of the EGF receptor and p53 mutations are mutually exclusive in the evolution of primary and secondary glioblastomas. Brain Pathol. 1996;6:217–223. doi: 10.1111/j.1750-3639.1996.tb00848.x. discussion 223–224. [DOI] [PubMed] [Google Scholar]
  • 36.Wong AJ, Ruppert JM, Bigner SH, Grzeschik CH, Humphrey PA, Bigner DS, Vogelstein B. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc Natl Acad Sci USA. 1992;89:2965–2969. doi: 10.1073/pnas.89.7.2965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Shinojima N, Tada K, Shiraishi S, Kamiryo T, Kochi M, Nakamura H, Makino K, Saya H, Hirano H, Kuratsu J, Oka K, Ishimaru Y, Ushio Y. Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res. 2003;63:6962–6970. [PubMed] [Google Scholar]
  • 38.Mischel PS, Cloughesy TF. Targeted molecular therapy of GBM. Brain Pathol. 2003;13:52–61. doi: 10.1111/j.1750-3639.2003.tb00006.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Albert L, Karsy M, Murali R, Jhanwar-Uniyal M. Inhibition of mTOR Activates the MAPK Pathway in Glioblastoma Multiforme. Cancer Genomics Proteomics. 2009;6:255–261. [PubMed] [Google Scholar]
  • 40.Stambolic V, Suzuki A, de la Pompa JL, Brothers GM, Mirtsos C, Sasaki T, Ruland J, Penninger JM, Siderovski DP, Mak TW. Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell. 1998;95:29–39. doi: 10.1016/s0092-8674(00)81780-8. [DOI] [PubMed] [Google Scholar]
  • 41.Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834–838. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
  • 42.Hurley JB, Simon MI, Teplow DB, Robishaw JD, Gilman AG. Homologies between signal transducing G proteins and ras gene products. Science. 1984;226:860–862. doi: 10.1126/science.6436980. [DOI] [PubMed] [Google Scholar]
  • 43.Moodie SA, Willumsen BM, Weber MJ, Wolfman A. Complexes of Ras.GTP with Raf-1 and mitogen-activated protein kinase kinase. Science. 1993;260:1658–1661. doi: 10.1126/science.8503013. [DOI] [PubMed] [Google Scholar]
  • 44.Thomas SM, DeMarco M, D’Arcangelo G, Halegoua S, Brugge JS. Ras is essential for nerve growth factor- and phorbol ester-induced tyrosine phosphorylation of MAP kinases. Cell. 1992;68:1031–1040. doi: 10.1016/0092-8674(92)90075-n. [DOI] [PubMed] [Google Scholar]
  • 45.Warner LC, Hack N, Egan SE, Goldberg HJ, Weinberg RA, Skorecki KL. RAS is required for epidermal growth factor-stimulated arachidonic acid release in rat-1 fibroblasts. Oncogene. 1993;8:3249–3255. [PubMed] [Google Scholar]
  • 46.Arvanitis D, Malliri A, Antoniou D, Linardopoulos S, Field JK, Spandidos DA. Ras p21 expression in brain tumors: elevated expression in malignant astrocytomas and glioblastomas multiforme. In Vivo. 1991;5:317–321. [PubMed] [Google Scholar]
  • 47.Blumenstock M, Prosenc N, Patt S, Pfanne K, Drum F, Cervos-Navarro J. In contrast to EGFr gene overexpression, H-ras gene expression decreases in human gliomas. Anti Cancer Res. 1991;11:1353–1357. [PubMed] [Google Scholar]
  • 48.Tsurushima H, Tsuboi K, Yoshii Y, Ohno T, Meguro K, Nose T. Expression of N-ras gene in gliomas. Neurol Med Chir (Tokyo) 1996;36:704–708. doi: 10.2176/nmc.36.704. [DOI] [PubMed] [Google Scholar]
  • 49.Abal M, Planaguma J, Gil-Moreno A, Monge M, Gonzalez M, Baro T, Garcia A, Castellvi J, Ramon Y, Cajal S, Xercavins J, Alameda F, Reventos J. Molecular pathology of endometrial carcinoma: transcriptional signature in endometrioid tumors. Histol Histopathol. 2006;21:197–204. doi: 10.14670/HH-21.197. [DOI] [PubMed] [Google Scholar]
  • 50.Rahaman SO, Harbor PC, Chernova O, Barnett GH, Vogelbaum MA, Haque SJ. Inhibition of constitutively active Stat3 suppresses proliferation and induces apoptosis in glioblastoma multiforme cells. Oncogene. 2002;21:8404–8413. doi: 10.1038/sj.onc.1206047. [DOI] [PubMed] [Google Scholar]
  • 51.Bonni A, Sun Y, Nadal-Vicens M, Bhatt A, Frank DA, Rozovsky I, Stahl N, Yancopoulos GD, Greenberg ME. Regulation of gliogenesis in the central nervous system by the JAK-STAT signaling pathway. Science. 1997;278:477–483. doi: 10.1126/science.278.5337.477. [DOI] [PubMed] [Google Scholar]
  • 52.Yoshimatsu T, Kawaguchi D, Oishi K, Takeda K, Akira S, Masuyama N, Gotoh Y. Non-cell-autonomous action of STAT3 in maintenance of neural precursor cells in the mouse neocortex. Development (Cambridge, England) 2006;133:2553–2563. doi: 10.1242/dev.02419. [DOI] [PubMed] [Google Scholar]
  • 53.Rajan P, McKay RD. Multiple routes to astrocytic differentiation in the CNS. J Neurosci. 1998;18:3620–3629. doi: 10.1523/JNEUROSCI.18-10-03620.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.de la Iglesia N, Konopka G, Puram SV, Chan JA, Bachoo RM, You MJ, Levy DE, Depinho RA, Bonni A. Identification of a PTEN-regulated STAT3 brain tumor suppressor pathway. Genes Dev. 2008;22:449–462. doi: 10.1101/gad.1606508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Li M, Zhang Y, Liu Z, Bharadwaj U, Wang H, Wang X, Zhang S, Liuzzi JP, Chang S-M, Cousins RJ, Fisher WE, Brunicardi FC, Logsdon CD, Chen C, Yao Q. Aberrant expression of zinc transporter ZIP4 (SLC39A4) significantly contributes to human pancreatic cancer pathogenesis and progression. Proc Natl Acad Sci USA. 2007;104:18636–18641. doi: 10.1073/pnas.0709307104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang Y, Bharadwaj U, Logsdon CD, Chen C, Yao Q, Li M. ZIP4 regulates pancreatic cancer cell growth by activating IL-6/STAT3 pathway through zinc finger transcription factor CREB. Clin Cancer Res. 2010;16:1423–1430. doi: 10.1158/1078-0432.CCR-09-2405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.O’Reilly SM, Newlands ES, Glaser MG, Brampton M, Rice-Edwards JM, Illingworth RD, Richards PG, Kennard C, Colquhoun IR, Lewis P. Temozolomide: a new oral cytotoxic chemotherapeutic agent with promising activity against primary brain tumours. Eur J Cancer. 1993;29A:940–942. doi: 10.1016/s0959-8049(05)80198-4. [DOI] [PubMed] [Google Scholar]
  • 58.Jacinto FV, Esteller M. MGMT hypermethylation: a prognostic foe, a predictive friend. DNA Repair (Amst) 2007;6:1155–1160. doi: 10.1016/j.dnarep.2007.03.013. [DOI] [PubMed] [Google Scholar]
  • 59.Prados MD, Chang SM, Butowski N, DeBoer R, Parvataneni R, Carliner H, Kabuubi P, Ayers-Ringler J, Rabbitt J, Page M, Fedoroff A, Sneed PK, Berger MS, McDermott MW, Parsa AT, Vandenberg S, James CD, Lamborn KR, Stokoe D, Haas-Kogan DA. Phase II study of erlotinib plus temozolomide during and after radiation therapy in patients with newly diagnosed glioblastoma multiforme or gliosarcoma. J Clin Oncol. 2009;27:579–584. doi: 10.1200/JCO.2008.18.9639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.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]
  • 61.Raizer JJ, Abrey LE, Lassman AB, Chang SM, Lamborn KR, Kuhn JG, Yung WK, Gilbert MR, Aldape KA, Wen PY, Fine HA, Mehta M, DeAngelis LM, Lieberman F, Cloughesy TF, Robins HI, Dancey J, Prados MD North American Brain Tumor Consortium. A phase I trial of erlotinib in patients with nonprogressive glioblastoma multiforme postradiation therapy, and recurrent malignant gliomas and meningiomas. Neuro Oncol. 2010;12:87–94. doi: 10.1093/neuonc/nop017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Raizer JJ, Abrey LE, Lassman AB, Chang SM, Lamborn KR, Kuhn JG, Yung WK, Gilbert MR, Aldape KA, Wen PY, Fine HA, Mehta M, DeAngelis LM, Lieberman F, Cloughesy TF, Robins HI, Dancey J, Prados MD North American Brain Tumor Consortium. A phase II trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy. Neuro Oncol. 2010;12:95–103. doi: 10.1093/neuonc/nop015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.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 astrocytoma: 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]
  • 64.Morse MA, Lyerly HK, Clay TM, Abdel-Wahab O, Chui SY, Garst J, Gollob J, Grossi PM, Kalady M, Mosca PJ, Onaitis M, Sampson JH, Seigler HF, Toloza EM, Tyler D, Vieweg J, Yang Y. Immunotherapy of surgical malignancies. Curr Probl Surg. 2004;41:15–132. doi: 10.1016/S0011384003001321. [DOI] [PubMed] [Google Scholar]
  • 65.Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov. 2006;5:671–688. doi: 10.1038/nrd2062. [DOI] [PubMed] [Google Scholar]
  • 66.Cloughesy TF, Yoshimoto K, Nghiemphu P, Brown K, Dang J, Zhu S, Hsueh T, Chen Y, Wang W, Youngkin D, Liau L, Martin N, Becker D, Bergsneider M, Lai A, Green R, Oglesby T, Koleto M, Trent J, Horvath S, Mischel PS, Mellinghoff IK, Sawyers CL. Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN-deficient glioblastoma. PLoS Med. 2008;5:e8. doi: 10.1371/journal.pmed.0050008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chang SM, Wen P, Cloughesy T, Greenberg H, Schiff D, Conrad C, Fink K, Robins HI, De Angelis L, Raizer J, Hess K, Aldape K, Lamborn KR, Kuhn J, Dancey J, Prados MD North American Brain Tumor Consortium, National Cancer Institute. Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme. Invest New Drugs. 2005;23:357–361. doi: 10.1007/s10637-005-1444-0. [DOI] [PubMed] [Google Scholar]
  • 68.Galanis E, Buckner JC, Maurer MJ, Kreisberg JI, Ballman K, Boni J, Peralba JM, Jenkins RB, Dakhil SR, Morton RF, Jaeckle KA, Scheithauer BW, Dancey J, Hidalgo M, Walsh DJ North Central Cancer Treatment Group. Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a North Central Cancer Treatment Group Study. J Clin Oncol. 2005;23:5294–5304. doi: 10.1200/JCO.2005.23.622. [DOI] [PubMed] [Google Scholar]
  • 69.Sarkaria JN, Galanis E, Wu W, Peller PJ, Giannini C, Brown PD, Uhm JH, McGraw S, Jaeckle KA, Buckner JC. North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys. 2011;81:468–475. doi: 10.1016/j.ijrobp.2010.05.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.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]
  • 71.Trinh XB, Tjalma WA, Vermeulen PB, Van den Eynden G, Van der Auwera I, Van Laere SJ, Helleman J, Berns EM, Dirix LY, van Dam PA. The VEGF pathway and the AKT/mTOR/p70S6K1 signalling pathway in human epithelial ovarian cancer. Br J Cancer. 2009;100:971–978. doi: 10.1038/sj.bjc.6604921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zhang H, Bajraszewski N, Wu E, Wang H, Moseman AP, Dabora SL, Griffin JD, Kwiatkowski DJ. PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. J Clin Invest. 2007;117:730–738. doi: 10.1172/JCI28984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Los M, Roodhart JM, Voest EE. Target practice: lessons from phase III trials with bevacizumab and vatalanib in the treatment of advanced colorectal cancer. Oncologist. 2007;12:443–450. doi: 10.1634/theoncologist.12-4-443. [DOI] [PubMed] [Google Scholar]
  • 74.Aoki K, Yoshida T, Matsumoto N, Ide H, Hosokawa K, Sugimura T, Terada M. Gene therapy for peritoneal dissemination of pancreatic cancer by liposome-mediated transfer of herpes simplex virus thymidine kinase gene. Hum Gene Ther. 1997;8:1105–1113. doi: 10.1089/hum.1997.8.9-1105. [DOI] [PubMed] [Google Scholar]
  • 75.Pope WB, Lai A, Nghiemphu P, Mischel P, Cloughesy TF. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 2006;66:1258–1260. doi: 10.1212/01.wnl.0000208958.29600.87. [DOI] [PubMed] [Google Scholar]
  • 76.Pope WB, Kim HJ, Huo J, Alger J, Brown MS, Gjertson D, Sai V, Young JR, Tekchandani L, Cloughesy T, Mischel PS, Lai A, Nghiemphu P, Rahmanuddin S, Goldin J. Recurrent glioblastoma multiforme: ADC histogram analysis predicts response to bevacizumab treatment. Radiology. 2009;252:182–189. doi: 10.1148/radiol.2521081534. [DOI] [PubMed] [Google Scholar]
  • 77.Sathornsumetee S, Sathornsumetee S, Desjardins A, Vredenburgh JJ, McLendon RE, Marcello J, Herndon JE, Mathe A, Hamilton M, Rich JN, Norfleet JA, Gururangan S, Friedman HS, Reardon DA. Phase II trial of bevacizumab and erlotinib in patients with recurrent malignant glioma. Neuro Oncol. 2010;12:1300–1310. doi: 10.1093/neuonc/noq099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Raizer JJ, Grimm S, Chamberlain MC, Nicholas MK, Chandler JP, Muro K, Dubner S, Rademaker AW, Renfrow J, Bredel M. A phase 2 trial of single-agent bevacizumab given in an every-3-week schedule for patients with recurrent high-grade gliomas. Cancer. 2010;116:5297–5305. doi: 10.1002/cncr.25462. [DOI] [PubMed] [Google Scholar]
  • 79.Batchelor TT, Duda DG, di Tomaso E, Ancukiewicz M, Plotkin SR, Gerstner E, Eichler AF, Drappatz J, Hochberg FH, Benner T, Louis DN, Cohen KS, Chea H, Exarhopoulos A, Loeffler JS, Moses MA, Ivy P, Sorensen AG, Wen PY, Jain RK. 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;28:2817–2823. doi: 10.1200/JCO.2009.26.3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kar R, Sen S, Singh A, Sharma H, Kumar S, Gupta SD, Singh N. Role of apoptotic regulators in human epithelial ovarian cancer. Cancer Biol Ther. 2007;6:1101–1105. doi: 10.4161/cbt.6.7.4329. [DOI] [PubMed] [Google Scholar]
  • 81.Hideshima T, Catley L, Yasui H, Ishitsuka K, Raje N, Mitsiades C, Podar K, Munshi NC, Chauhan D, Richardson PG, Anderson KC. Perifosine, an oral bioactive novel alkylphospholipid, inhibits Akt and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells. Blood. 2006;107:4053–4062. doi: 10.1182/blood-2005-08-3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Reardon DA, Vredenburgh JJ, Desjardins A, Peters K, Gururangan S, Sampson JH, Marcello J, Herndon JE, 2nd, McLendon RE, Janney D, Friedman AH, Bigner DD, Friedman HS. Effect of CYP3A-inducing anti-epileptics on sorafenib exposure: results of a phase II study of sorafenib plus daily temozolomide in adults with recurrent glioblastoma. J Neurooncol. 2011;101:57–66. doi: 10.1007/s11060-010-0217-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Razis E, Selviaridis P, Labropoulos S, Norris JL, Zhu MJ, Song DD, Kalebic T, Torrens M, Kalogera-Fountzila A, Karkavelas G, Karanastasi S, Fletcher JA, Fountzilas G. Phase II study of neoadjuvant imatinib in glioblastoma: evaluation of clinical and molecular effects of the treatment. Clin Cancer Res. 2009;15:6258–6266. doi: 10.1158/1078-0432.CCR-08-1867. [DOI] [PubMed] [Google Scholar]
  • 84.Reardon DA, Dresemann G, Taillibert S, Campone M, van den Bent M, Clement P, Blomquist E, Gordower L, Schultz H, Raizer J, Hau P, Easaw J, Gil M, Tonn J, Gijtenbeek A, Schlegel U, Bergstrom P, Green S, Weir A, Nikolova Z. Multicentre phase II studies evaluating imatinib plus hydroxyurea in patients with progressive glioblastoma. Br J Cancer. 2009;101:1995–2004. doi: 10.1038/sj.bjc.6605411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tsunetsugu-Yokota Y, Morikawa Y, Isogai M, Kawana-Tachikawa A, Odawara T, Nakamura T, Grassi F, Autran B, Iwamoto A. Yeast-derived human immunodeficiency virus type 1 p55(gag) virus-like particles activate dendritic cells (DCs) and induce perforin expression in Gag-specific CD8(+) T cells by cross-presentation of DCs. J Virol. 2003;77:10250–10259. doi: 10.1128/JVI.77.19.10250-10259.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Gerstner ER, Eichler AF, Plotkin SR, Drappatz J, Doyle CL, Xu L, Duda DG, Wen PY, Jain RK, Batchelor TT. Phase I trial with biomarker studies of vatalanib (PTK787) in patients with newly diagnosed glioblastoma treated with enzyme inducing anti-epileptic drugs and standard radiation and temozolomide. J Neurooncol. 2011;103:325–332. doi: 10.1007/s11060-010-0390-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Graff JR, McNulty AM, Hanna KR, Konicek BW, Lynch RL, Bailey SN, Banks C, Capen A, Goode R, Lewis JE, Sams L, Huss KL, Campbell RM, Iversen PW, Neubauer BL, Brown TJ, Musib L, Geeganage S, Thornton D. The protein kinase Cbeta-selective inhibitor, Enzastaurin (LY317615.HCl), suppresses signaling through the AKT pathway, induces apoptosis, and suppresses growth of human colon cancer and glioblastoma xenografts. Cancer Res. 2005;65:7462–7469. doi: 10.1158/0008-5472.CAN-05-0071. [DOI] [PubMed] [Google Scholar]
  • 88.Wick W, Puduvalli VK, Chamberlain MC, van den Bent MJ, Carpentier AF, Cher LM, Mason W, Weller M, Hong S, Musib L, Liepa AM, Thornton DE, Fine HA. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol. 2010;28:1168–1174. doi: 10.1200/JCO.2009.23.2595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Strong JE, Coffey MC, Tang D, Sabinin P, Lee PW. The molecular basis of viral oncolysis: usurpation of the Ras signaling pathway by reovirus. Embo J. 1998;17:3351–3362. doi: 10.1093/emboj/17.12.3351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Forsyth P, Roldán G, George D, Wallace C, Palmer CA, Morris D, Cairncross G, Matthews MV, Markert J, Gillespie Y, Coffey M, Thompson B, Hamilton M. A phase I trial of intratumoral administration of reovirus in patients with histologically confirmed recurrent malignant gliomas. Mol Ther. 2008;16:627–632. doi: 10.1038/sj.mt.6300403. [DOI] [PubMed] [Google Scholar]
  • 91.Heimberger AB, Archer GE, Mitchell AD, Bigner DD, Schmittling RJ, Herndon JE, II, Davis T, Friedman HS, Keler T, Reardon DA, Sampson JH. Epidermal growth factor receptor variant III (EGFRvIII) vaccine (CDX-110) in GBM. ASCO Meeting Abstracts. 2009;27:2021. [Google Scholar]
  • 92.Iwamoto Fabio M, Lamborn Kathleen R, Robins H. Ian, Mehta Minesh P, Chang Susan M, Butowski Nicholas A, DeAngelis Lisa M, Abrey Lauren E, Zhang Wei-Ting, Prados Michael D, Howard A. Phase II trial of pazopanib (GW786034), an oral multi-targeted angiogenesis inhibitor, for adults with recurrent glioblastoma (North American Brain Tumor Consortium Study 06-02) Neuro-Oncology. 2010;12:855–861. doi: 10.1093/neuonc/noq025. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES