Quick viewing(Text Mode)

Chemotherapy for Glioblastoma: Current Treatment and Future Perspectives for Cytotoxic and Targeted Agents

Chemotherapy for Glioblastoma: Current Treatment and Future Perspectives for Cytotoxic and Targeted Agents

ANTICANCER RESEARCH 29: 5171-5184 (2009)

Review for Glioblastoma: Current Treatment and Future Perspectives for Cytotoxic and Targeted Agents

G. MINNITI1,2, R. MUNI1, G. LANZETTA2, P. MARCHETTI3 and R. MAURIZI ENRICI1

Departments of 1Radiotherapy Oncology and 3Medical Oncology, Sant’ Andrea Hospital University La Sapienza, Rome; 2Department of Neuroscience, NEUROMED Institute, Pozzilli (IS), Italy

Abstract. Glioblastoma is the most frequent and review summarizes the current results with cytotoxic and devastating primary malignant brain tumor in adults. targeted molecular agents in glioblastoma and the Surgery followed by standard radiotherapy with development of new chemoradiation strategies under concomitant and adjuvant chemotherapy with temozolomide evaluation to increase their effectiveness. is the standard of care in patients with glioblastoma, however the prognosis remains poor with a median survival Approximately 176,000 cases of central nervous system in the range of 12-15 months. Common genetic (CNS) cancer are diagnosed per year worldwide, with an abnormalities in glioblastoma are associated with aberrant estimated annual mortality of 128,000 (1). Gliomas are the activation or suppression of cellular signal transduction most frequent intracranial tumor and account for more than pathways and resistance to radiation and chemotherapy. 50% of all primary brain tumors, with glioblastoma Special attention has been focused on targets such as (GBM) being by far the most common and aggressive (2, epidermal receptor, vascular endothelial 3). Treatment of GBM includes surgery, radiotherapy (RT) , platelet-derived growth factor and chemotherapy. Currently, surgery followed by standard receptor, and on pathways such as the phosphatidylinositol- RT with concomitant and adjuvant chemotherapy with 3kinase/Akt/mammalian target of rapamycin and temozolomide is the standard of care in patients with GBM Ras/Raf/mitogen-activated protein-kinase pathways. Several aged <70 years, however the prognosis remains poor, with signal transduction inhibitors have been examined in a median survival of 12-15 months (4). Clearly, there is a preclinical and clinical malignant glioma trials, including desperate need for more effective therapies for patients antiangiogenic agents (, enzastaurin), and with GBM. inhibitors of receptor tyrosine Overexpression, activation, and dysregulation of various kinase ( and ), mammalian target of membrane receptors, signaling pathways, and other factors rapamycin (, ) and occur frequently in GBM (5-7). Therapeutic approaches (cilengitide). Although preliminary clinical results of the targeting these molecules, including inhibitors of epidermal use of targeted agents have not translated into significantly growth factor receptor (EGFR), vascular endothelial growth better survival, more recent phase II trials are exploring the factor receptor (VEGFR), platelet-derivated growth factor combination of multitargeted drugs with cytotoxic receptor (PDGFR), and mammalian target of rapamycin chemotherapy and radiotherapy in order to overcome the (mTOR) have been examined in preclinical and clinical trials resistance of tumors to single-agent targeted therapies. This for malignant glioma. The efficacy of these agents as monotherapies has been modest, at best; however, new multitargeted kinase inhibitors and combinations of single- targeted kinase inhibitors in combination with radiation and Correspondence to: Dr. Giuseppe Minniti, Sant’ Andrea Hospital, cytotoxic chemotherapy will likely play an increasing role in Department of Radiotherapy Oncology, Via di Grottarossa 1035, the management of GBM and several randomized/prospective 00189 Roma, Italy. Tel: +39 0633776034, Fax: +39 0633776608, e-mail: [email protected] studies are ongoing. We have performed a review of the current results with chemotherapy in malignant glioma and Key Words: Glioblastoma, radiotherapy, chemotherapy, temozolomide, of the new treatment strategies that may improve the , review. prognosis for these patients.

0250-7005/2009 $2.00+.40 5171 ANTICANCER RESEARCH 29: 5171-5184 (2009)

Current Standard Therapy for GBM in the range of 7-9 months and 10-14 months, respectively, which compare favourably with series using RT alone (12-14). Temozolomide is a second-generation imidazotetrazine In 32 elderly patients of 70 years or older treated with derivative, which exerts its cytoxic effects by methylation of standard RT and temozolomide given concomitantly and specific DNA sites. The methylation of the O6 position of adjuvantly, the median survival was 10.8 months and the guanine in DNA is usually regarded as the most critical. median progression-free survival was 6.7 months (12). The 12- Temozolomide is taken orally and is absorbed rapidly, with a month survival and progression-free survival rates were 36% nearly complete bioavailability after oral administration, and 16%, respectively. Longer survival has been reported for acrosses the brain-blood barrier, and achieves high elderly patients who had MGMT promoter methylated (14). concentration in the cerebrospinal fluid. Prolonged schedules with higher cumulative dose of The recently published randomized European and Canadian temozolomide have been explored in order to maximize trial (EORTC 26981/22981-NCIC) (4) has clearly demonstrated MGMT depletion in cancer cells and possibly to increase that the addition of temozolomide to RT, followed by 6 monthly antitumor efficacy (15-17). Regimens using temozolomide cycles of temozolomide provides significant survival benefit with administered 1 week on/1 week off or 21 days on/7 days off minimal additional toxicity in patients with GBM. The reported in patients with recurrent malignant glioma result in a more median survival was 14.6 months with RT plus temozolomide protracted MGMT depletion in blood cells and possibly in and 12.1 months with RT alone, with respective 2-year survival brain tumor tissue. rates of 27% and 10%. The 2-year survival rate improved from Wick et al. (16) reported on 90 patients with recurrent 10.4% for RT alone to 26.5% in the RT plus temozolomide malignant gliomas treated with temozolomide (150 mg/m2) group. A recent up-date of the trial reported the final results, with administered 1 week on and 1 week off, showing a promising a median follow-up of more than 5 years (8). Overall survival median progression-free survival of 24 weeks, being 43.8% at 6 was 27.2% at 2 years, 16.0% at 3 years, 12.1% at 4 years, and months. The RTOG 052/EORTC 26052/220535 trial, led by the 9.8% (6.4-14.0) at 5 years in RT plus temozolomide group, Radiation Therapy Oncology Group (RTOG) along with the versus 10.9%, 4.4%, 3.0%, and 1.9% in the radiotherapy alone European Organisation for Research and Treatment of Cancer group (p<0.0001). A benefit of combined therapy was recorded (EORTC) and the North Central Cancer Treatment Group, in all clinical prognostic subgroups, including younger age, which has enrolled 1,150 patients (closed in June 2008), is extent of resection, and good performance status. Methylation of evaluating if a dose-intensity schedule of temozolomide (75 to the O6-methylguanine-DNA-methyltransferase (MGMT) 100 mg/m2 days 1 to 21 every 28 days for 12 cycles maximum) promoter was the strongest predictor of outcome and benefit improves survival compared with standard chemotherapy from temozolomide chemotherapy. regimen. Currently, in the absence of robust data, the use of The cytotoxic effect of temozolomide is correlated to the dose-intense temozolomide schedules as alternatives to standard intracellular levels of MGMT. MGMT is a critical DNA repair regimens are not recommended in patients with GBM. protein that protects tumor cells against alkylating In summary, temozolomide given concomitantly and chemotherapeutic agents, tranferring the methyl group to an adjuvantly with RT after surgical resection represents the internal cysteine acceptor residue. High levels of MGMT standard of care in patients with GBM. The epigenetic activity in tumor tissue are associated with resistance to silencing of MGMT by promoter methylation is correlated temozolomide (9). Thus, epigenetic silencing of the MGMT with improved survival in patients treated with DNA-repair gene by promoter methylation results in decreased temozolomide. Although the prolonged exposure of cancer levels of MGMT and may enhance temozolomide sensitivity, cells to temozolomide may represent a promising strategy to whereas unmethylated MGMT promoter is associated with overcome resistance mediated by MGMT, at present, the potential temozolomide resistance (8, 10, 11). Hegi et al. (10) clinical relevance of the use of a dose-dense temozolomide reported a survival advantage in GBM patients with schedule remains to be proven. methylated MGMT promoter treated by temozolomide and standard radiotherapy, whereas GBM patients with Targeted Therapies unmethylated MGMT promoter did not seem to have a survival benefit by chemoradiation combination. The 2-year survival GBM is characterized by several aberrantly activated signaling rates for patients with unmethylated and those with methylated pathways. Several growth factor receptors, such as epidermal MGMT promoter treated with standard RT and temozolomide growth factor receptor (EGFR), vascular endothelial growth were 14% and 46%, respectively. factor receptor (VEGFR) and platelet-derivated growth factor The use of standard or hypofractionated RT plus receptor (PDGFR), are overexpressed, amplified and/or concomitant and/or adjuvant temozolomide has been recently mutated, leading to uncontrolled cell proliferation, extended to elderly people with GBM. The reported median angiogenesis, migration, survival and differentiation (18, 19) progression-free survival and median survival durations were (Figure 1). Thus, several targeted agents are being developed

5172 Minniti et al: Chemotherapy for Glioblastoma (Review)

Figure 1. Schematic representation of main oncogenetic signaling pathways and site of action of molecular targeted therapies. EGFR: epidermal growth factor receptor; PDGFR: platelet-derived growth factor receptor; VEGFR, vascular endothelial growth factor receptor; PLC, phospholipase C; PKC, protein kinase C; P13K, phosphatidylinositol 3’-kinase; PTEN, phosphatase and tensin homolog; Akt, v-akt murine thymoma viral oncogene homolog; mTOR, mammalian-target of rapamycin.

as potential inhibitors of growth factor receptor transduction EGFR Inhibitors pathways and angiogenesis (Table I). The major challenge in the use of targeted therapies is the identification of the optimal EGFR is a 170-kDa receptor member of the therapeutic targets, and to select new agents which can translate ErbB family which consists of four distinct receptors: to survival benefit for patients with GBM. Currently, the use HER1/EGFR (epidermal growth factor receptor), HER2, HER3 of small molecule tyrosine kinases inhibitors (TKIs) and and HER4. It is formed of three major domains: the monoclonal antibodies against EGFR and VEGFR have been extracellular domain, the transmembrane domain and the evaluated in phase II clinical trials as main anti-growth factor cytoplasmatic domain, which harbors the tyrosine kinase receptor strategies. Other potentially useful agents for the activity. Phosphorylation of the tyrosine kinase domain activates treatment of GBM are represented by the PDGFR-, several signaling pathways, such as the phosphatidyl-inositol 3’- mammalian target of rapamycin (mTOR) and integrin kinase (PI3K)/Akt/mTOR, and Ras/mitogen-activated protein inhibitors. A summary of main recent phase II clinical trials of kinase (MAPK) (20, 21). Activation of EGFR pathways results targeted therapies alone or in combination with other treatment in several biological processes, including cell proliferation, modalities in GBM are shown in Table II. angiogenesis, migration, survival and differentiation.

5173 ANTICANCER RESEARCH 29: 5171-5184 (2009)

Table I. Main molecular targeted agents used in the treatment of developed in order to block the EGFR signaling pathways, glioblastoma. including monoclonal antibodies and small molecule TKIs. Preclinical studies with the murine humanized monoclonal Agent Mechanism of action antibody (Erbitux, Merck & Co) have shown that Inhibitors of EGFR inhibition leads to while reducing EGFR proliferation as well as VEGF expression in GBM cells (29). Cetuximab EGFR blocker (monoclonal antibody) An additive cytotoxic effect can be observed in different Gefitinib EGFR TKI GBM cell lines by the addition of cetuximab to Erlotinib EGFR TKI EGFR, Erb-2 TKI radiochemotherapy with temozolomide (30). Currently a Erb-1, Erb-2, Erb-3, Erb-4 TKI one-armed single-center phase I/II trial is evaluating the Pelitinib Irreversible EGFR inhibitor safety and efficacy of combined radiotherapy with BIBW-2992 EGFR and HER2 inhibitor temozolomide and cetuximab as first-line treatment for PDGFR patients with primary GBM (31). mesylate PDGFR TKI, c-Kit, Bcr-Abl Tandutinib PDGFR, FLT3 (FMS-like tyrosine kinase-3), Gefitinib (Iressa, AstraZeneca) and erlotinib (Tarceva, Osi c-Kit inhibitor Pharmaceuticals) are two oral, small TKI molecules that VEGFR have been evaluated in several phase I and II studies in Bevacizumab VEGFR blocker (monoclonal antibody) patients with GBM. Phase I and II studies of gefitinib and VEGF trap Pan VEGF blocker erlotinib used as single agents adjuvant to RT have shown no Vatalinib VEGFR-1, VEGFR-2, PDGFR, c-Kit TKI EGFR, HER2 inhibitor major improvements in survival (32-38). In a phase II trial Cediranib Pan-VEGFR, PDGFR, C- TKI that included 53 patients with recurrent GBM, gefitinib was VEGFR, PDGFR, c-Kit TKI well tolerated, however, it resulted in a modest progression- VEGFR-1, VEGFR-2, VEGFR-3, PDGFR, free and median survival of 8.1 and 39.4 weeks respectively c-Kit TKI (33); similar results have been shown by others (29, 31, 32). mTOR Sirolimus mTOR inhibitor Phase II trials of erlotinib single-agent therapy in recurrent Temsirolimus mTOR inhibitor GBM have demonstrated a similar median progression-free Everolimus mTOR inhibitor survival of only 2-3 months, with response rates of 6-25% AP23573 mTOR inhibitor (37-39). In a recent randomized phase II trial (EORTC Brain PKC Tumor Group Study 26034) of erlotinib versus temozolomide Enzastaurin PKC inhibitor RAF-MEK-ERK or carmustine in 110 patients with recurrent GBM, the 6- Tipifarnib Farnesyltransferase inhibitor month progression free survival rate was 11.4% in the Lonafarnib Farnesyltransferase inhibitor erlotinib arm and 24% in the control arm (40). The reported Inhibitor of farnesyltransferase I and clinical evidence clearly suggests that the use of EGFR VEGFR and PDGFR TK inhibitors erlotinib and gefitinib as single agents in patients with progressive GBM is not beneficial. Cilengitide ανβ3 and ανβ5 integrin inhibitor There is little evidence that erlotinib and gefitinib may be EGFR, Epidermal growth factor receptor; VEGF, vascular endothelial associated with survival advantages in a subgroup of patients growth factor; VEGFR, vascular endothelial growth factor receptor; with GBM who have overexpressed, amplified, or mutated PDGFR, platelet-derived growth factor receptor; PKC, protein kinase EGFR in association with intact PTEN or unphosphorylated C; TKI, tyrosine kinase inhibitor. Akt (41, 42). Mellinghoff et al. (42) reviewed the molecular characteristics of 26 patients with malignant gliomas treated with EGFR TKIs. Coexpression of EGFRvIII and PTEN, a EGFR amplification and/or overexpression occur in tumor-suppressor protein that inhibits the PI3K signaling approximately half of GBM cases (22, 23). EGFRvIII is a pathway, was significantly associated with better clinical mutant EGFR variant characterized by deletion of the response to EGFR TKIs. Loss of PTEN may promote extracellular ligand-binding domain, which results in a resistance to EGFR kinase inhibitors. In contrast, Lassman constitutionally active receptor and is reported in 20-40% of et al. (43) found no correlation between sensitivity to GBMs (24, 25). All these alterations are responsible for the erlotinib or gefitinib and changes in expression, activation of EGFR downstream signaling pathways, amplification, phosphorylation, downstream signaling of promote the oncogenic process, and are associated with a EGFR, and genomic changes. negative prognosis (26, 27). Moreover, in the clinical setting, More recently, phase I/II studies testing the combination EGFR overexpression in GBM has been associated with of erlotinib in association with RT and/or temozolomide resistance to radiation therapy (28). Therefore, a large showed conflicting results (44-46). Using this regimen, number of potentially therapeutic targets have been Prados et al. (46) showed a median progression-free survival

5174 Minniti et al: Chemotherapy for Glioblastoma (Review)

Table II. Main recent phase II clinical trials of targeted therapies alone or in combination with cytotoxic chemotherapy and/or radiotherapy in newly diagnosed and recurrent glioblastoma.

Study Treatment Patients Dose of Tumor Response Median Median (n) target agent (CR+PR) PFS OS (mg/day) % (months) (months)

EGFR inhibitors Lieberman et al. (32) Gefitinib 38 500-1000** R 13 2 NA Rich et al. (33) Gefitinib 53 500-1000** R 0 2 10 Uhm et al. (34) Gefitinib 98 500-1000** N NA 7 11 Franceschi et al. (35) Gefitinib 28* 250 R 0 14.3% at 6 months Raizer et al. (37) Erlotinib 30 150 R 0 3 NA Vogelbaum et al. (37) Erlotinib 31 150 R 26 3 6 Cloughesy et al. (38) Erlotinib 48 150-500** R 8 2 NA Brown et al. (44) Erlotinib +TMZ + RT 97 150 N NA NA 15.3 Prados et al. (45) Erlotinib +TMZ + RT 65 100-300** N NA 8.2 19.3 van den Bent et al. (39) Erlotinib vs. TMZ or BCNU 110 100-300** R NA 11.4 vs. 24% NA at 6 months

PDGF inhibitors Reardon et al. (53) Imatinib + hydroxyurea 33 400-1000** R 9 4.5 27% at 6 months Wen et al. (51) Imatinib 34 600-800** R 6 NA NA Raymond et al. (52) Imatinib 51 600-800** R 6 16% at 6 months NA

VEGF inhibitors Vredenburgh et al. (63) Bevacizumab + irinotecan 35 10-15 mg/kg q14-21 R 100 46% at 6 months 77% at 6 months Bokstein et al. (66) Bevacizumab + irinotecan 20 5 mg/kg q14 R 47 25% at 6 months 45% at 6 months Narayana et al. (70) Bevacizumab + TMZ + RT 15 10 mg/kg q14 N 93 59% at 1 year 87% at 12 months Norden et al. (68) Bevacizumab 55 10 mg/kg q14 R 34 42% at 6 months NA Kreisl et al. (64) Bevacizumab + irinotecan*** 48 10 mg/kg q14 R 35^ 4 7.7 Conrad et al. (79) Vatalanib 47 150-2.000 R 4 NA NA Batchelor et al. (82) Cediranib 30 45 qd R 56 3.7 7.7 mTor inhibitors Chang et al. (105) Temsirolimus 43 170-250 R 5 2.3 NA Galanis et al. (106) Temsirolimus 65 250 R 0 2.3 4

Protein kinase C, RAF-MEK- ERK, and integrin inhibitors Fine et al. (110) Enzastaurin versus 167/84 500 R NA 1.5 vs. 2.8 6.6 vs. 7.1 Cloughesy et al. (112) Tipifarnib 67 300-600 bid** R 7 12% at 6 months NA Reardon et al. (119) Cilengitide 81 500-2000 R NA 15% at 6 months 9.9 Stupp et al. (120) Cilengitide + TMZ + RT 52 500 mg i.v. 2 x/w N NA 65% at 6 months NA

TMZ, Temozolomide; RT, radiotherapy; NA, not available; CR, complete response; PR, partial response; PFS, progression-free survival; OS, overall survival. *Series includes some grade III gliomas; **based on enzyme-inducing antiepileptic drug (EIAED) therapy. ***bevacizumab was initially used as single agent and in combination with irinotecan at progression; ^no responses in pts treated with bev+irinotecan at progression.

of 8.2 months and median survival of 19.3 months. EGFRvIII plus wild-type PTEN compared with patients Compared with the EORTC 26981/22981-NCIC conducted without these genotypic features. Moreover, the regimen was by Stupp et al. (4), where patients were treated with RT and associated with significant toxicity, including two grade 5 temozolomide alone, the study showed survival benefit, toxicities (non-neutropenic pneumonias). To date, the especially in patients with MGMT promoter methylation and potential role of erlotinib in association with temozolomide PTEN positivity. By contrast, a recent study of the North and RT remains unclear. Central Cancer Treatment Group (45) using a similar New growth factor receptor inhibitors are emerging as regimen showed no significant additional benefit for erlotinib potential therapeutic agents in GBM. Lapatinib (Tykerb, when combined with RT and temozolomide, and no Glaxo Smith-Kline) is a dual EGFR and ErbB-2 inhibitor differences in survival for patients characterized by that has shown some activity in the treatment of solid

5175 ANTICANCER RESEARCH 29: 5171-5184 (2009) tumors, especially of advanced or metastatic breast cancer patients with recurrent GBM, Raymond et al. (52) reported a (47). Canertinib (Pfizer, New York) is a pan-ErbB inhibitor 6-month progression-free survival of 16% with 3 patients which has shown potent and sustained inhibition of tyrosine who had an objective partial response. Imatinib mesylate kinase activity selective for erbB1, erbB2, erbB3, and erbB4 plus hydroxyurea is associated with better antitumor activity in human cancer cell lines (48). Treatment of athymic nude than imatinib given alone. At a median follow-up of 58 mice bearing xenografts of human SF-767 glioblastoma with weeks, Reardon et al. (53) reported a median progression- canertinib resulted in highly significant suppression of tumor free survival of 14.4 weeks and 6-month progression-free growth (49). However, no clinical data are available in survival of 26% in 33 patients with recurrent GBM. Nine patients with high-grade gliomas. percent of patients achieved radiographic response, and 42% In summary, the patterns of response observed in patients of patients achieved stable disease. The most common treated with EGFR TKIs are disappointing. Despite EGFR toxicities included grade 3 neutropenia in 16% of patients, activating several signaling pathways, such as PI3K/Akt/ thrombocytopenia in 6% of patients, and edema in 6% of mTOR, and Ras/MAPK, it is possible that other growth factor patients. In a phase I study of 55 patients with GBM, the and signaling pathways play a greater role in maintaining the combination of imatinib plus temozolomide was well transformed phenotype of GBM. Erlotinib in association with tolerated (54). Grade 3 or 4 hematological toxicities occurred RT and temozolomide has led to modest or no survival benefit in fewer than 10% of patients. The most common compared to RT plus temozolomide alone, and currently its use nonhematological toxicities included grade 2 nausea/emesis is not advisable in clinical practice. New strategies include the and fatigue in 5% of patients. Overall survival and combination of EGFR TKIs with targeted agents able to inhibit progression-free survival were 45.1 and 26.6 weeks, with a other signaling pathways, and phase II trials testing the 6-month survival of 53%. In summary, in the dose range of association with temsilorus and sorafenib are ongoing 600 to 1,000 mg/d, single-agent imatinib is well tolerated but (http://clinicaltrials.gov/ct2/show/NCT00474786). has limited antitumor activity in patients with recurrent GBM, and available data do not support its use in clinical Platelet-derived Growth Factor Inhibitors practice. Combination regimens incorporating imatinib are encouraging and phase II trials have been planned. Imatinib mesylate (Gleevec, Novartis) is an inhibitor of the kinase activity of multiple growth factor receptors, including VEGF/VEGFR Inhibitors PDGFR, c-Kit, and Bcr-Abl. The PDGF family consists of four members, PDGF-A, -B, -C and -D, which signal Vascular proliferation, or neoangiogenesis, is a distinct through the alpha and beta PDGF receptor (PDGFRα and histopathological characteristic of GBM and is correlated PDGFRβ) tyrosine kinases. Binding of ligands induces with prognosis (55-57). VEGF is a key factor involved in the dimerization and cross-tyrosine phosphorylation of the angiogenetic process that can elicit several responses such as intracellular domain. This results in the activation of PI3K, endothelial cell proliferation, extracellular matrix degradation, MAPK, Jak family kinase, Src family kinase, and cell migration, and expression of other proangiogenic factors phospholipase C-gamma (PLC-γ) signal transduction (matrix metalloproteinase-1, urokinase-type plasminogen pathways. Overexpression of alpha subtype receptor activator and its receptor, plasminogen activator inhibitor-1). (PDGFRα) has been reported in approximately one third of VEGF expression is stimulated by hypoxia, acidosis, and primary glioblastoma (50). c-Kit is a many growth factors as EGF, PDGF, that acts similarly to EGFR and PDGFR and is (HGF), c-Kit and their downstream signaling pathways overexpressed in GBM. c-Kit consists of an extracellular (PI3K-Akt, Ras-MAPK). The main receptors involved in domain, a transmembrane segment, a juxtamembrane relaying VEGF-A signaling are VEGFR-1 and VEFGR-2. segment, and a protein kinase domain. Binding of stem cell The rationale for anti-VEGF/VEGFR therapies has been factor to Kit results in receptor dimerization and activation supported by several in vitro and in vivo studies (58, 59). of several signal transduction pathways including Akt, Src Bevacizumab (Avastin, Genentech) is a humanized murine family kinases, PI3K, PLC-gamma, and Ras/MAPK. monoclonal antibody that selectively blocks VEGF, preventing Imatinib mesylate used as single agent in the dose range the activation of VEGF receptor tyrosine kinases VEGFR1 and of 600 to 1,000 mg/d is generally well tolerated but shows VEGFR2 (60). In patients with metastatic and limited activity in patients with recurrent GBM (51, 52). In a recurrent non-small cell (NSCLC), bevacizumab small phase I/II study of the North American Brain Tumor in combination with conventional chemotherapy resulted in Consortium (51) including 34 patients with recurrent GBM, longer survival (61, 62). Phase II studies in patients with the 6-month progression-free survival was 3%, with only 2 recurrent GBM using bevacizumab alone or in association with and 6 patients who had partial response and stable disease, irinotecan have been reported (63-69) (Table II). Vredenburgh respectively. In a recent phase II EORTC study including 51 et al. (63) reported on 35 patients with recurrent GBM treated

5176 Minniti et al: Chemotherapy for Glioblastoma (Review) with bevacizumab plus irinotecan. Twenty-three patients models, alone or in combination with RT (71-78), and are received bevacizumab at 10 mg/kg plus irinotecan every 2 currently undergoing clinical evaluation in GBM. weeks and 12 patients received bevacizumab 15 mg/kg every Vatalanib is an oral VEGFR-1 and VEGFR-2 TKI that is 21 days and irinotecan on days 1, 8, 22, and 29 for a total of 6 able to inhibit VEGFR, PDGFR, c-Kit and colony- cycles. The 6-month progression-free survival and overall stimulating factor 1 receptor (CSF1R). It has been recently survival rates were 46% and 77%, respectively. Fifty-seven tested in two phase I/II studies given alone or in combination percent of patients had at least a partial radiological response. with cytotoxic chemotherapy (79-80). In a study of 47 One patient developed a CNS hemorrhage, and 4 patients patients with recurrent GBM treated with vatalanib as single developed thromboembolic complications. Kreisl et al. (64) agent, a partial response was shown in 4% and stability of reported on 48 patients with recurrent GBM treated with disease in 56% of patients (79). Reardon et al. (80) reported bevacizumab followed by bevacizumab plus irinotecan at the results of vatalanib in association with either progression of tumor. The median survival and progression-free temozolomide or lomustine in 51 patients. The progression- survival were 31 weeks and 16 weeks, with a respective 6- free survival was 16 weeks in vatalanib plus temozolomide month survival and progression-free survival of 57% and 29%. group and 10 weeks in vatalanib plus lomustine group. A Thirty-five percent of patients achieved a radiographic recent phase I trial from the same authors has shown that response. In contrast, amongst 19 patients treated with vatalanib at doses up to 1000 mg twice-a-day combined with bevacizumab plus irinotecan at progression, the median imatinib and hydroxyurea were well tolerated (81). progression-free survival was only 30 days, with no objective Cediranib is an oral pan-VEGFR TKI that also inhibits radiographic responses. Thromboembolic events (12.5%), PDGFR and C-Kit. Batchelor et al. (82) reported the results hypertension (12.5%), hypophosphatemia (6%), and of cediranib used as single agent in 16 patients with recurrent thrombocytopenia (6%) were the most common drug- GBM, showing a promising median overall survival of 7 associated adverse events. Similar 6-month progression-free months and median progression-free survival of 3.5 months. and overall survival, and discontinuation of steroids for Magnetic resonance imaging (MRI) imaging showed a recurrent GBM to a bevacizumab-based treatment have been significant decrease in tumor enhancement in more than 50% reported by others (65-68), suggesting that bevacizumab of patients. Contrast enhancement decreased after a single improves local control and survival in this patient population. dose of cediranib in all patients, and this was associated with Based on this clinical evidence, more recently the US Food and a significant reduced vasogenic edema. Toxicity was modest Drug Administration (FDA) Oncologic Drugs Advisory and included diarrhea, dysphonia, and hypertension. A phase Committee (ODAC) has approved the use bevacizumab as a III randomized trial comparing the efficacy of cediranib single agent for patients with GBM, with progressive disease monotherapy or the combination of cediranib with lomustine following prior therapy. to the efficacy of lomustine alone is planned (http:// The feasibility of using bevacizumab with chemoradiation in clinicaltrials.gov/ct2/show/NCT00777153). the primary management of GBM has been tested in few studies Sunitinib and sorafenib are two multitarget TKIs that have (69, 70). Narayana et al. (70) treated 15 patients with standard demonstrated potent antitumor and antiangiogenic activity in RT and bevacizumab at the dose of 10 mg/kg on days 14 and 28 preclinical models (72, 75, 77, 78). Sunitinib is a small, and daily temozolomide (75 mg/m2). Subsequently, orally bioavailable molecule which is able to inhibit PDGFR- bevacizumab at the same dose was continued every 2 weeks α and PDGFR-β, as well as VEGFR-1 and -2, and c-KIT. with temozolomide (150 mg/m2) for 12 months. Radiographic Sunitinib has demonstrated efficacy and tolerability in responses were noted in 13 of 14 assessable patients (93%). several phase II/III clinical trials involving patients with One-year progression-free survival and overall survival rates gastrointestinal stromal tumor, metastatic renal cell were 59% and 87%, respectively. Four patients had grade III/IV carcinoma, and is currently tested in breast cancer and nonhematologic toxicities. These positive outcomes have led to NSCLC (83-85). There are no clinical data in patients with the initiation of a new phase III trial (RTOG-0825) testing GBM; however, phase I/II studies of Sunitinib as single- concurrent chemoradiation and adjuvant temozolomide plus agent (http://clinicaltrials.gov/ct2/show/NCT00713388) or in bevacizumab versus conventional chemoradiation and adjuvant combination with irinotecan (http://clinicaltrials.gov/ temozolomide in patients with newly diagnosed GBM. archive/NCT00611728) are ongoing. Sorafenib is one of the Several new antiangiogenic inhibitors have been recently most promising agents targeting PDGFR, VEGFR, and Raf developed, including vatalanib (Novartis Pharmaceuthical), and has been recently approved by the Food and Drug cediranib (Recentin, Astra Zeneca), sunitinib (Sutent, Pfizer), Administration for the treatment of renal cell carcinoma (86- sorafenib (Nexavar, Bayer Pharmaceuticals), vandetanib 88). Strategies to antagonize PKC and ERK pathways in (Zactima, Astra Zeneca), and VEGF trap (, combination seem to be an attractive approach to enhance Regeneron Pharmaceuticals). These targeted agents have therapeutic efficacy in human glioma cells also, and a phase demonstrated antitumor activity in preclinical glioma II evaluation of sorafenib in glioma as a single agent and in

5177 ANTICANCER RESEARCH 29: 5171-5184 (2009) combination with temozolomide or erlotinib is ongoing These evidence has provided the rationale for clinical (http://clinicaltrials.gov/ct2/show/NCT00445588). studies of mTOR inhibitors in GBM. Vandetanib is an inhibitor of VEGFR-2 tyrosine kinase with Temsirolimus (Torisel, Wyeth Pharmaceutical), everolimus additional effects on VEGFR-3 and EGFR (89, 90) that has (Certican, Novartis Pharmaceutical) and AP23573 (ARIAD shown significant antitumor effects in combination with Pharmaceutical) are three synthetic analogs of rapamycin radiotherapy in rat glioma models (91). Phase I/II studies are (sirolimus) currently being tested as potential targeted drugs exploring the use of vandetanib alone or in combination with in GBM. Results of phase II studies using temsirolimus as chemoradiation in patients with malignant glioma (http:// single agent found only a limited activity in recurrent GBM clinicaltrials.gov/ct2/show/NCT00821080 and /NCT00613223), (105, 106). More recently, based on evidence of the however, currently no clinical data are available. synergism between mTOR inhibitors and EGFR TKI (107), VEGF Trap is a 110 kDa soluble protein that is able to ongoing trials have been exploring such a combination (108, bind circulating VEGF, inhibiting its activity (71), and to 109). A phase I study found no significant pharmacokinetics potentiate RT in preclinical GBM xenografts (92). De Groot interaction between sirolimus and gefitinib, with partial et al. (93) reported the results of a phase II trial of VEGF radiological response and stable disease that were seen only Trap monotherapy for 48 patients with recurrent GBM and in 2 and 13 of 34 patients, respectively (108). Similarly, a anaplastic glioma at first relapse. Response rates were 50% study combining gefitinib and everolimus failed to find for the anaplastic glioma group and 30% for the glioblastoma survival benefit of this regimen in comparison with historical group. Grade 3 toxicity occurred in 40% of patients and controls, although a radiological response occurred in about included fatigue, hypertension, hand-foot syndrome, one third of patients (109). lymphopenia, thrombosis and proteinuria, causing the Despite these preliminary disappointing results, several discontinuation of therapy in 25% of patients. studies are exploring the safety and efficacy of mTOR In summary, VEGF inhibitors target one important inhibitors in combination with other targeted agents and/or pathway in brain tumors. Bevacizumab represents the first chemoirradiation. Ongoing phase I/II studies are assessing antiangiogenic agent approved for use in malignant glioma. the maximum tolerable dose and the efficacy of temsirolimus In addition to bevacizumab, there is an array of inhibitors of plus sorafenib (http://www.cancer.gov/clinicaltrials/NCCTG- VEGF, VEGFR, and other relevant targets that could be N0572), temsirolimus plus sorafenib and erlotinib (http:// effective in selected patients with GBM. Combining these clinicaltrials.gov/ct2/show/NCT00335764), and temsirolimus antiangiogenic agents with conventional chemotherapy or or everolimus plus irradiation (http://groups.eortc.be/ radiation is a promising antitumor strategy that is currently brain/html/trials/26082-22081) or chemoradiation with being investigated in several phase II clinical trials with the temozolomide (http://clinicaltrials.gov/ct2/show/NCT005 intent to produce a synergistic antitumor effect and a survival 53150). benefit for patients with GBM, targeting both the tumor cell In summary, although data strongly support the view of and tumor vascular compartment. Certainly, an improved the PTEN/PI3K/AKT pathway as an important target for understanding of resistance to antiangiogenic therapy is drugs, current clinical results on the use of mTOR inhibitors necessary to identify effective strategies for the patients that remains disappointing. A new generation of trials is seeking progress despite treatment. to define whether the combination of two or more targeted drugs together with RT and cytotoxic chemotherapy can Mammalian Target of Rapamycin Inhibitors overcome tumor resistance.

Overactivation of the PI3K/Akt/mTOR pathway seems to Protein Kinase C, RAF-MEK-ERK, play an important role in gliomagenesis (94). Combined and Integrin Inhibitors activation of Ras and Akt leads to the formation of GBM in mice (95). In human GBM, Akt is activated in Enzastaurin is an oral serine/threonine kinase inhibitor that approximately 70% of these tumors, in association with selectively inhibits PKC-β, a mediator of VEGF intracellular loss of PTEN and/or activation of EGFR and PDGFR signaling. Unfortunately, a phase III study comparing tyrosine kinases. Alterations of PTEN expression are lomustine with enzastaurin in recurrent GBM was present in 20-40% of GBM (96, 97), and have been discontinued at the first interim analysis of 266 patients due associated with a worse prognosis, although conflicting to lack of survival benefit of enzastaurin over the control results have been reported (97-99). There is emerging group (109). As for other targeted agents that failed to show evidence suggesting that mTOR is a critical downstream tumor activity when used as single agents, combinations of component in PTEN/Akt signaling (100-103), and enzastaurin with RT (110-111) or temozolomide pharmacological inactivation of mTOR reduces neoplastic (http://www.eortc.be/protoc/26054) are currently being proliferation and tumor size in PTEN-deficient mice (104). investigated in phase I/II studies.

5178 Minniti et al: Chemotherapy for Glioblastoma (Review)

The RAF-MEK-ERK signal transduction pathway is an Conclusion important mediator of dysregulated glioma cell proliferation and angiogenesis. Tipifarnib (Zarnestra, Johnson and The current standard treatment for GBM is chemoradiotherapy Johnson), lonarfanib (Sarasar, Shering-Plough), and with temozolomide following surgical resection. Identification sorafenib (Nexavar, Bayer) may inactivate RAS by inhibiting of many molecular genetic and signal transduction pathways farnesyltransferase. A phase II study conducted in 22 patients involved in oncogenesis have yielded many targeted drugs that with recurrent malignant glioma treated with tipifarnib (300 are currently undergoing clinical evaluation in GBM. Currently, mg twice a day for 21 days every 4 weeks) showed modest several drugs have been tested, including EGFR TKIs (gefitinib evidence of activity, with a 6-month progression-free survival and erlotinib), antiangiogenic agents (bevacizumab, enzastaurin), of 12% (112). Combination of these agents in association and inhibitors of mTOR (temsirolimus, everolimus) and integrin with radiotherapy and chemotherapy has been reported in (cilengitide). Although bevacizumab has been recently approved few studies (113, 114). In a phase I study of 13 patients with for use as a single agent for patients with GBM, with progressive GBM, tipifarnib (200 mg/day) concurrent with standard RT disease, most of the molecular targeted therapy phase II clinical was well tolerated and resulted in a median survival of 12 trials in GBM have not translated into significant survival months (113). Lonafarnib in combination with temozolomide advantages. Better molecular characterization of GBM could resulted in a 6-month progression-free survival of 38% in 23 avoid negative results in large clinical trials enrolling patients with recurrent GBM (114). Phase I/II studies on the heterogeneous cohorts of patients, and potentially allow the combination of tipifarnib, lonarfanib, and sorafenib in design of “individualized” therapies based on the genetic and association with RT, temozolomide and other targeted agents molecular characteristics of each tumor. New chemotherapeutic (http://clinicaltrials.gov/ct2/show) are ongoing, however no strategies are represented by the combination of multitargeted clinical results are currently available. drugs with cytotoxic chemotherapy and radiotherapy in order to Integrins are heterodimeric cell surface adhesion receptors overcome tumor resistance. Randomized clinical trials remain that consist of two noncovalently associated alpha and beta the gold standard for the evaluation of new targeted agents. Most subunits, and function as receptors for extracellullar matrix multicenters randomized European (EORTC) and American proteins such as fibronectin, laminins, collagens, and (RTOG, NABTG) clinical trials are testing these new agents in vitronectin (115). Ligand binding to the extracelluar domain combination with standard chemoradiotherapy (RT plus of integrin receptors results in receptor activation and in the concomitant and adjuvant temozolomide) versus standard transduction of signals essential for cell adhesion, migration, chemoradiotherapy alone. This is important because all the proliferation, differentiation, and survival (116). The integrins patients will be treated appropriately. Hopefully, results from ανβ3 and ανβ5 appear to be particularly important in the these randomized trials and the development of new process of angiogenesis and are expressed in a variety of chemoradiation strategies will translate into real benefit for malignancies, including gliomas. The critical role of integrins patients with GBM. in angiogenesis and association with tumor progression make them an attractive target for anticancer therapy. Cilengitide References (EMD 121974), a cyclic Arg-Gly-Glu (RGD) peptide is a potent and selective inhibitor of the integrins ανβ3 and ανβ5 1 Parkin DM: Global cancer statistics in the year 2000. Lancet (115-118). A phase II study has been conducted in 81 patients Oncol 2: 533-543, 2001. with recurrent GBM randomly assigned to receive either 500 2 Davis FG, Freels S, Grutsch J, Barlas S and Brem S: Survival or 2,000 mg of cilengitide twice weekly on a continuous basis rates in patients with primary malignant brain tumors stratified (119). Cilengitide was well tolerated, with no significant by patient age and tumor histological type: an analysis based toxicities observed in either arm. A modest antitumor activity on Surveillance, Epidemiology, and End Results (SEER) data, 1973-1991. J Neurosurg 88: 1-10, 1998. was observed in both treatment groups, with a 6-month 3 Minniti G, Brada M, Kleihues P and Ohgaki H: Gliomas. In: progression-free survival of 15% and a median overall Prognostic Factors in Cancer, 3rd edition (Gospodarowicz MK, survival of 9.9 months, which were more favorable among O’Sullivan B, Sobin LH (eds.). Hoboken, New Jersey, Wiley J patients treated with 2,000 mg. In a phase I/II trial of RT with & Sons, pp. 307-312, 2006. concomitant and adjuvant cilengitide and temozolomide, 4 Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Stupp et al. (120) reported a 6-month progression-free Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, survival of 69% and a 12-month survival of 67%, with no Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E and Mirimanoff RO; added toxicity. A phase III EORTC 26071-22072 study is European Organisation for Research and Treatment of Cancer testing cilengitide in combination with standard treatment Brain Tumor and Radiotherapy Groups; National Cancer (standard RT with concomitant and adjuvant temozolomide) Institute of Canada Clinical Trials Group: Radiotherapy plus versus standard treatment in patients with newly diagnosed concomitant and adjuvant temozolomide for glioblastoma. GBM with methylated MGMT promoter gene. N Engl J Med 352: 987-996, 2005.

5179 ANTICANCER RESEARCH 29: 5171-5184 (2009)

5 Kapoor GS and O’Rourke DM: Receptor tyrosine kinase alternating weekly regimen in patients with recurrent glioma. signaling in gliomagenesis: pathobiology and therapeutic J Clin Oncol 25: 3357-3361, 2007. approaches. Cancer Biol Ther 2: 330-342, 2003. 17 Neyns B, Chaskis C, Joosens E, Menten J, D’Hondt L, Branle 6 Kitange GJ, Templeton KL and Jenkins RB: Recent advances F, Sadones J and Michotte A: A multicenter cohort study of in the molecular genetics of primary gliomas. Curr Opin Oncol dose-dense temozolomide (21 of 28 days) for the treatment of 15: 197-203, 2003. recurrent anaplastic astrocytoma or oligoastrocytoma. Cancer 7 Louis DN: Molecular pathology of malignant gliomas. Annu Invest 26: 269-277, 2008. Rev Pathol 1: 97-117, 2006. 18 Sanson M, Thillet J and Hoang-Xuan K: Molecular changes in 8 Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, gliomas. Curr Opin Oncol 16: 607-613, 2004. Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau 19 de Groot JF and Gilbert MR: New molecular targets in P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K, malignant gliomas. Curr Opin Neurol 20: 712-718, 2007. Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, 20 Normanno N, De Luca A, Bianco C, Strizzi L, Mancino M, Lacombe D, Cairncross JG and Mirimanoff RO; European Maiello MR, Carotenuto A, De Feo G, Caponigro F and Organisation for Research and Treatment of Cancer Brain Salomon DS: Epidermal growth factor receptor (EGFR) Tumour and Radiation Oncology Groups; National Cancer signaling in cancer. Gene 366: 2-16, 2006. Institute of Canada Clinical Trials Group: Effects of 21 Burgess AW: EGFR family: structure physiology signalling and radiotherapy with concomitant and adjuvant temozolomide therapeutic targets. Growth Factors 26: 263-274, 2008. versus radiotherapy alone on survival in glioblastoma in a 22 Smith JS, Tachibana I, Passe SM, Huntley BK, Borell TJ, randomised phase III study: 5-year analysis of the EORTC- Iturria N, O’ Fallon JR, Schaefer PL, Scheithauer BW, James NCIC trial. Lancet Oncol 10: 434-439, 2009. CD, Buckner JC and Jenkins RB: PTEN mutation, EGFR 9 Baer JC, Freeman AA, Newlands ES, Watson AJ, Rafferty JA amplification, and outcome in patients with anaplastic and Margison GP: Depletion of O6-alkylguanine-DNA alkyl- astrocytoma and glioblastoma multiforme. J Natl Cancer Inst transferase correlates with potentiation of temozolomide and 93: 1246-1256, 2001. CCNU toxicity in human tumour cells. Br J Cancer 67: 1299- 23 Heimberger AB, Hlatky R, Suki D, Yang D, Weinberg J, Gilbert 1302, 1993. M, Sawaya R and Aldape K: Prognostic effect of epidermal 10 Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, growth factor receptor and EGFRvIII in glioblastoma Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, multiforme patients. Clin Cancer Res 11: 1462-1466, 2005. Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC 24 Frederick L, Wang XY, Eley G and James CD: Diversity and and Stupp R: MGMT gene silencing and benefit from frequency of epidermal growth factor receptor mutations in temozolomide in glioblastoma. N Engl J Med 352: 997-1003, human glioblastomas. Cancer Res 60: 1383-1387, 2000. 2005. 25 Aldape KD, Ballman K, Furth A, Buckner JC, Giannini C, 11 Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller M, Burger PC, Scheithauer BW, Jenkins RB and James CD: Mehta MP and Gilbert MR: Correlation of O6-methylguanine Immunohistochemical detection of EGFRvIII in high methyltransferase (MGMT) promoter methylation with clinical malignancy grade astrocytomas and evaluation of prognostic outcomes in glioblastoma and clinical strategies to modulate significance. J Neuropathol Exp Neurol 63: 700-707, 2004. MGMT activity. J Clin Oncol 26: 4189-4199, 2008. 26 Shinojima N, Tada K, Shiraishi S, Kamiryo T, Kochi M, 12 Minniti G, De Sanctis V, Muni R, Rasio D, Lanzetta G, Bozzao Nakamura H, Makino K, Saya H, Hirano H, Kuratsu J, Oka K, A, Osti MF, Salvati M, Valeriani M, Cantore GP and Maurizi Ishimaru Y and Ushio Y: Prognostic value of epidermal growth Enrici R: Hypofractionated radiotherapy followed by adjuvant factor receptor in patients with glioblastoma multiforme. chemotherapy with temozolomide in elderly patients with Cancer Res 63: 6962-6970, 2003. glioblastoma. J Neurooncol 91: 95-100, 2008. 27 Pelloski CE, Ballman KV, Furth AF, Zhang L, Lin E, Sulman 13 Minniti G, De Sanctis V, Muni, R Filippone F, Bozzao A, EP, Bhat K, McDonald JM, Yung WK, Colman H, Woo SY, Valeriani M, Osti MF, De Paula U, Lanzetta G, Tombolini V Heimberger AB, Suki D, Prados MD, Chang SM, Barker FG and Maurizi Enrici R: Radiotherapy plus concomitant and 2nd, Buckner JC, James CD and Aldape K: Epidermal growth adjuvant temozolomide for glioblastoma in elderly patients. factor receptor variant III status defines clinically distinct J Neurooncol 88: 97-103, 2008. subtypes of glioblastoma. J Clin Oncol 25: 2288-2294, 2007. 14 Brandes AA, Franceschi E, Tosoni A, Benevento F, Scopece L, 28 Barker FG 2nd, Simmons ML, Chang SM, Prados MD, Larson Mazzocchi V, Bacci A, Agati R, Calbucci F and Ermani M: DA, Sneed PK, Wara WM, Berger MS, Chen P, Israel MA and Temozolomide concomitant and adjuvant to radiotherapy in Aldape KD: EGFR overexpression and radiation response in elderly patients with glioblastoma: correlation with MGMT glioblastoma multiforme. Int J Radiat Oncol Biol Phys 51: 410- promoter methylation status. Cancer 115: 3512-3351, 2009. 418, 2001. 15 Brandes AA, Tosoni A, Cavallo G, Bertorelle R, Gioia V, 29 Eller JL, Longo SL, Hicklin DJ and Canute GW: Activity of Franceschi E, Biscuola M, Blatt V, Crinò L and Ermani M; anti-epidermal growth factor receptor monoclonal antibody GICNO: Temozolomide 3 weeks on and 1 week off as first-line C225 against glioblastoma multiforme. Neurosurgery 51: 1005- therapy for recurrent glioblastoma: phase II study from Gruppo 1013, 2002. Italiano Cooperativo di Neuro-Oncologia (GICNO). Br J 30 Combs SE, Schulz-Ertner D, Roth W, Herold-Mende C, Debus Cancer 95: 1155-1160, 2006. J and Weber KJ: In vitro responsiveness of glioma cell lines to 16 Wick A, Felsberg J, Steinbach JP, Herrlinger U, Platten M, multimodality treatment with radiotherapy, temozolomide, and Blaschke B, Meyermann R, Reifenberger G, Weller M and epidermal growth factor receptor inhibition with cetuximab. Int Wick W: Efficacy and tolerability of temozolomide in an J Radiat Oncol Biol Phys 68: 873-882, 2007.

5180 Minniti et al: Chemotherapy for Glioblastoma (Review)

31 Combs SE, Heeger S, Haselmann R, Edler L, Debus J and 42 Mellinghoff IK, Wang MY, Vivanco, I Haas-Kogan DA, Zhu S, Schulz-Ertner D: Treatment of primary glioblastoma multiforme Dia EQ, Lu KV, Yoshimoto K, Huang JH, Chute DJ, Riggs BL, with cetuximab, radiotherapy and temozolomide (GERT)-phase Horvath S, Liau LM, Cavenee WK, Rao PN, Beroukhim R, I/II trial: study protocol. BMC Cancer 6: 133, 2006. Peck TC, Lee JC, Sellers WR, Stokoe D, Prados M, Cloughesy 32 Lieberman FS, Cloughesy T, Fine H, Kuhn Lamborn JK, Malkin TF, Sawyers CL and Mischel PS: Molecular determinants of the M, Robbins HI, Yung WA, Wen P and Prados M: NABTC phase response of glioblastomas to EGFR kinase inhibitors. N Engl J I/II trial of ZD-1839 for recurrent malignant gliomas and Med 353: 2012-2024, 2005. unresectable meningiomas. J Clin Oncol ASCO Annual Meeting 43 Lassman AB, Rossi MR, Raizer JJ Abrey LE, Lieberman FS, Proc (Post-Meeting Edition) 22(Suppl): 1510, 2004. Grefe CN, Lamborn K, Pao W, Shih AH, Kuhn JG, Wilson R, 33 Rich JN, Reardon DA, Peery T, Dowell JM, Quinn JA, Penne Nowak NJ, Cowell JK, DeAngelis LM, Wen P, Gilbert MR, KL, Wikstrand CJ, Van Duyn LB, Dancey JE, McLendon RE, Chang S, Yung WA, Prados M and Holland EC: Molecular Kao JC, Stenzel TT, Ahmed Rasheed BK, Tourt-Uhlig SE, study of malignant gliomas treated with epidermal growth Herndon JE 2nd, Vredenburgh JJ, Sampson JH, Friedman AH, factor receptor inhibitors: tissue analysis from North American Bigner DD and Friedman HS: Phase II trial of gefitinib in Brain Tumor Consortium Trials 01-03 and 00-01. Clin Cancer recurrent glioblastoma. J Clin Oncol 22: 133-142, 2004. Res 11: 7841-7850, 2005. 34 Uhm JH, Ballman KV, Giannini, Krauss JC, Buckner JC, James 44 Prados MD, Lamborn KR, Chang S Burton E, Butowski N, D, Scheithauer BW, O’ Fallon JR and Jaeckle KA: Phase II Malec M, Kapadia A, Rabbitt J, Page MS, Fedoroff A, Xie D study of ZD1839 in patients with newly diagnosed grade 4 and Kelley SK: Phase 1 study of erlotinib HCl alone and astrocytoma. J Clin Oncol ASCO Annual Meeting Proc combined with temozolomide in patients with stable or 22(Suppl): 1505, 2004. recurrent malignant glioma. Neuro Oncol 8: 67-78, 2006. 35 Franceschi E, Lo nardi S, Tosoni A, Grosso D, Scopece L, 45 Brown PD, Krishnan S, Sarkaria JN, Wu W, Jaeckle KA, Uhm Berzioli C, Urbini B, Cavallo G, Crinò L and Brandes A: JH, Geoffroy FJ, Arusell R, Kitange G, Jenkins RB, Kugler JW, ZD1839 (Iressa) treatment for adult patients with progressive Morton RF, Rowland KM Jr, Mischel P, Yong WH, Scheithauer high-grade gliomas (HGG): an open label, single-arm, phase II BW, Schiff D, Giannini C and Buckner JC: North Central study of the Gruppo Italiano Cooperativo di Neuro-Oncologia Cancer Treatment Group Study N0177: Phase I/II trial of (GICNO). J Clin Oncol ASCO Annual Meeting Proc 23(Suppl): erlotinib and temozolomide with radiation therapy in the 1564, 2005. treatment of newly diagnosed glioblastoma multiforme: North 36 Chakravarti A, Seiferheld W, Robins HI, Guha A, Brachman D, Central Cancer Treatment Group Study N0177. J Clin Oncol Curran W, Choucair A and Mehta M: An update of phase I data 26: 5603-5609, 2008. from RTOG 0211: A phase I/II clinical study of gefitinib+ 46 Prados MD, Chang SM, Butowski N, DeBoer R, Parvataneni R, radiation for newly-diagnosed glioblastoma (GBM) patients. J Carliner H, Kabuubi P, Ayers-Ringler J, Rabbitt J, Page M, Clin Oncol ASCO Annual Meeting Proc 24(Suppl): 1507, 2006. Fedoroff A, Sneed PK, Berger MS, McDermott MW, Parsa AT, 37 Raizer JJ, Abrey LE, Wen P, Cloughesy T, Robins IA, Fine HA, Vandenberg S, James CD, Lamborn KR, Stokoe D and Haas- Lieberman F, Puduvalli VK, Fink KL and Prados M: A phase Kogan DA: Phase II study of erlotinib plus temozolomide II trial of erlotinib (OSI-774) in patients (pts) with recurrent during and after radiation therapy in patients with newly malignant gliomas (MG) not on EIAEDs. J Clin Oncol ASCO diagnosed glioblastoma multiforme or gliosarcoma. J Clin Annual Meeting Proc (Post-Meeting Edition) 22(Suppl): 1502, Oncol 27: 579-584, 2009. 2004. 47 Gomez HL, Doval DC, Chavez MA, Ang PC, Aziz Z, Nag S, 38 Vogelbaum MA, Peereboom D, Stevens G, Barnett G and Brewer Ng C, Franco SX, Chow LW, Arbushites MC, Casey MA, C: Phase II trial of the EGFR tyrosine kinase inhibitor erlotinib Berger MS, Stein SH and Sledge GW: Efficacy and safety of for single agent therapy of recurrent glioblastoma multiforme: lapatinib as first-line therapy for ErbB2-amplified locally Interim results. J Clin Oncol ASCO Annual Meeting Proc advanced or metastatic breast cancer. J Clin Oncol 26: 2999- 22(Suppl): 1558, 2004. 3005, 2008. 39 Cloughesy T, Yung A, Vrendenberg J, Aldape K, Eberhard D, 48 Allen LF, Eiseman IA and Fry DW: CI-1033, an irreversible Prados M, Vandenberg S, Klencke B and Mischel P: Phase II pan-erbB receptor inhibitor and its potential application for the study of erlotinib in recurrent GBM: Molecular predictors of treatment of breast cancer. Semin Oncol 30: 65-78, 2003. outcome. J Clin Oncol ASCO Annual Meeting Proc 22(Suppl): 49 Christensen JG, Vincent PW, Klohs WD, Fry DW, Leopold WR, 1507, 2005. Elliott WL: Plasma vascular endothelial growth factor and 40 van den Bent MJ, Brandes AA, Rampling R, Kouwenhoven -8 as biomarkers of antitumor efficacy of a MC, Kros JM, Carpentier AF, Clement PM, Frenay M, prototypical erbB family tyrosine kinase inhibitor. Mol Cancer Campone M, Baurain JF, Armand JP, Taphoorn MJ, Tosoni A, Ther 4: 938-947, 2005. Kletzl H, Klughammer B, Lacombe D and Gorlia T: 50 Joensuu H, Puputti M, Sihto H, Tynninen O and Nupponen NN: Randomized phase II Trial of erlotinib versus temozolomide or Amplification of genes encoding KIT, PDGFRα and VEGFR2 carmustine in recurrent glioblastoma: EORTC Brain Tumor receptor tyrosine kinases is frequent in glioblastoma Group Study 26034. J Clin Oncol 27: 1268-1274, 2009. multiforme. J Pathol 207: 224-231, 2005. 41 Haas-Kogan DA, Prados MD, Tihan T, Eberhard DA, Jelluma 51 Wen PY, Yung WK, Lamborn KR, Dahia PL, Wang Y, Peng B, N, Arvold ND, Baumber R, Lamborn KR, Kapadia A, Malec Abrey LE, Raizer J, Cloughesy TF, Fink K, Gilbert M, Chang M, Berger MS and Stokoe D: Epidermal growth factor receptor, S, Junck L, Schiff D, Lieberman F, Fine HA, Mehta M, protein kinase B/Akt, and glioma response to erlotinib. J Natl Robins HI, DeAngelis LM, Groves MD, Puduvalli VK, Levin Cancer Inst 97: 880-887, 2005. V, Conrad C, Maher EA, Aldape K, Hayes M, Letvak L,

5181 ANTICANCER RESEARCH 29: 5171-5184 (2009)

Egorin MJ, Capdeville R, Kaplan R, Murgo AJ, Stiles C and and Fine HA: Phase II trial of single-agent bevacizumab Prados MD: Phase I/II study of imatinib mesylate for followed by bevacizumab plus irinotecan at tumor progression recurrent malignant gliomas: North American Brain Tumor in recurrent glioblastoma. J Clin Oncol 27: 740-745, 2009. Consortium Study 99-08. Clin Cancer Res 12: 4899-4907, 65 Raval S, Hwang S and Dorsett L: Bevacizumab and irinotecan 2006. in patients (pts) with recurrent glioblastoma multiforme (GBM). 52 Raymond E, Brandes AA, Dittrich C Fumoleau P, Coudert B, J Clin Oncol ASCO Annual Meeting Proc Part I 25(Suppl): Clement PM, Frenay M, Rampling R, Stupp R, Kros JM, 2078, 2007. Heinrich MC, Gorlia T, Lacombe D and van den Bent MJ; 66 Bokstein F, Shpigel S and Blumenthal DT: Treatment with European Organisation for Research and Treatment of Cancer bevacizumab and irinotecan for recurrent high-grade glial Brain Tumor Group Study: Phase II study of imatinib in tumors. Cancer 112: 2267-2273, 2008. patients with recurrent gliomas of various histologies: a 67 Vredenburgh JJ, Desjardins A, Herndon JE 2nd, Dowell JM, European Organisation for Research and Treatment of Cancer Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan Brain Tumor Group Study J Clin Oncol 26: 4659-4665, 2008. S, Wagner M, Bigner DD, Friedman AH and Friedman HS: 53 Reardon DA, Egorin MJ, Quinn, JA Rich JN, Gururangan S, Phase II trial of bevacizumab and irinotecan in recurrent Vredenburgh JJ, Desjardins A, Sathornsumetee S, Provenzale malignant glioma. Clin Cancer Res 13: 1253-1259, 2007. JM, Herndon JE 2nd, Dowell JM, Badruddoja MA, McLendon 68 Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R, RE, Lagattuta TF, Kicielinski KP, Dresemann G, Sampson JH, Ross GL, Ciampa AS, Ebbeling LG, Levy B, Drappatz J, Kesari Friedman AH, Salvado AJ and Friedman HS: Phase II study of S and Wen PY: Bevacizumab for recurrent malignant gliomas: imatinib mesylate plus hydroxyurea in adults with recurrent efficacy, toxicity, and patterns of recurrence. Neurology 70: glioblastoma multiforme. J Clin Oncol 23: 9359-9368, 2005. 779-787, 2008. 54 Reardon DA, Desjardins A, Vredenburgh JJ, Sathornsumetee S, 69 Lai A, Filka E, McGibbon B, Nghiemphu PL, Graham C, Yong Rich JN, Quinn JA, Lagattuta TF, Egorin MJ, Gururangan S, WH, Mischel P, Liau LM, Bergsneider M, Pope W, Selch M McLendon R, Herndon JE 2nd, Friedman AH, Salvado AJ and and Cloughesy T: Phase II pilot study of bevacizumab in Friedman HS: Safety and pharmacokinetics of dose-intensive combination with temozolomide and regional radiation therapy imatinib mesylate plus temozolomide: phase 1 trial in adults for up-front treatment of patients with newly diagnosed with malignant glioma. Neuro Oncol 10: 330-340, 2008. glioblastoma multiforme: interim analysis of safety and 55 Plate KH, Breier G, Weich HA and Risau W: Vascular tolerability. Int J Radiat Oncol Biol Phys 71: 1372-1380, 2008. endothelial growth factor is a potential tumour angiogenesis 70 Narayana A, Golfinos JG, Fischer I, Raza S, Kelly P, Parker E, factor in human gliomas in vivo. Nature 359: 845-848, 1992. Knopp EA, Medabalmi P, Zagzag D, Eagan P and Gruber ML: 56 Kleihus P and Cavenee WK. Pathology and Genetics of Feasibility of using bevacizumab with radiation therapy and Tumours of the Nervous System. IARC Press, Lyon, 2000. temozolomide in newly diagnosed high-grade glioma. Int J 57 Reijneveld JC, Voest EE and Taphoorn MJ: Angiogenesis in Radiat Oncol Biol Phys 72: 383-389, 2008. malignant primary and metastatic brain tumors. J Neurol 247: 71 Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell 597-608, 2000. M, Boland P, Leidich R, Hylton D, Burova E, Ioffe E, Huang T, 58 Chaudhry IH, O’Donovan DG, Brenchley PE, Reid H and Radziejewski C, Bailey K, Fandl JP, Daly T, Wiegand SJ, Roberts IS: Vascular endothelial growth factor expression Yancopoulos GD and Rudge JS: VEGF-Trap: a VEGF blocker correlates with tumour grade and vascularity in gliomas. with potent antitumor effects. Proc Natl Acad Sci USA 99: Histopathology 39: 409-415, 2001. 11393-11398, 2002. 59 Longo R, Sarmiento R, Fanelli M, Capaccetti B, Gattuso D and 72 Schueneman AJ, Himmelfarb E, Geng L, Tan J, Donnelly E, Gasparini G: Anti-angiogenic therapy: rationale, challenges and Mendel D, McMahon G and Hallahan DE: SU11248 maintenance clinical studies. Angiogenesis 5: 237-256, 2002. therapy prevents tumor regrowth after fractionated irradiation of 60 Ferrara N, Hillan KJ, Gerber HP and Novotny W: Discovery murine tumor models. Cancer Res 63: 4009-4016, 2003. and development of bevacizumab, an anti-VEGF antibody for 73 Sandström M, Johansson M, Andersson U, Bergh A, treating cancer. Nat Rev Drug Discov 3: 391-400, 2004. Bergenheim AT and Henriksson R: The tyrosine kinase 61 Hurwitz H, Fehrenbacher L, Novotny, Cartwright T, Hainsworth inhibitor ZD6474 inhibits tumour growth in an intracerebral rat J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara glioma model. Br J Cancer 91: 1174-1180, 2004. N, Fyfe G, Rogers B, Ross R and Kabbinavar F: Bevacizumab 74 Damiano V, Melisi D, Bianco C, Raben D, Caputo R, Fontanini plus irinotecan, fluorouracil, and leucovorin for metastatic G, Bianco R, Ryan A, Bianco AR, De Placido S, Ciardiello F colorectal cancer. N Engl J Med 350: 2335-2342, 2004. and Tortora G: Cooperative antitumor effect of multitargeted 62 Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati kinase inhibitor ZD6474 and ionizing radiation in glioblastoma. A, Lilenbaum R and Johnson DH: Paclitaxel-carboplatin alone Clin Cancer Res 11: 5639-5644, 2005. or with bevacizumab for non-small cell lung cancer. N Engl J 75 de Boüard S and Guillamo JS: Angiogenesis and anti-angiogenic Med 355: 2542-2550, 2006. strategies for glioblastoma. Bull Cancer 92: 360-372, 2005. 63 Vredenburgh JJ, Desjardins A, Herndon JE 2nd Marcello J, 76 Rich JN, Sathornsumetee S, Keir ST, Kieran MW, Laforme A, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan Kaipainen A, McLendon RE, Graner MW, Rasheed BK, Wang S, Sampson J, Wagner M, Bailey L, Bigner DD, Friedman AH L, Reardon DA, Ryan AJ, Wheeler C, Dimery I, Bigner DD and and Friedman HS: Bevacizumab plus irinotecan in recurrent Friedman HS: ZD6474, a novel tyrosine kinase inhibitor of glioblastoma multiforme. J Clin Oncol 25: 4722-4729, 2007. vascular endothelial growth factor receptor and epidermal 64 Kreisl TN, Kim L, Moore K Duic P, Royce C, Stroud I, Garren growth factor receptor, inhibits tumor growth of multiple N, Mackey M, Butman JA, Camphausen K, Park J, Albert PS nervous system tumors. Clin Cancer Res 11: 8145-8157, 2005.

5182 Minniti et al: Chemotherapy for Glioblastoma (Review)

77 Jane EP, Premkumar DR and Pollack IF: Co-administration of HL, Graham GA, Hughes GD, Thomas AP, Stokes ES, Curry sorafenib with rottlerin potently inhibits cell proliferation and B, Richmond GH, Wadsworth PF, Bigley AL and Hennequin migration in human malignant glioma cells. J Pharmacol Exp LF: ZD6474 inhibits vascular endothelial growth factor Ther 319: 1070-1080, 2006. signaling, angiogenesis, and tumor growth following oral 78 de Boüard S, Herlin P, Christensen JG, Lemoisson E, administration. Cancer Res 62: 4645-4655, 2002. Gauduchon P, Raymond E and Guillamo JS: Antiangiogenic 90 Ciardiello F, Caputo R, Damiano V, Caputo R, Troiani T, and anti-invasive effects of sunitinib on experimental human Vitagliano D, Carlomagno F, Veneziani BM, Fontanini G, glioblastoma. Neuro Oncol 9: 412-423, 2007. Bianco AR and Tortora G: Antitumor effects of ZD6474, a 79 Conrad C, Friedman H, Reardon D, Provenzale J, Jackson E, small molecule vascular endothelial growth factor receptor Serajuddin H, Laurent D, Chen B and Yung WKA: A phase I/II tyrosine kinase inhibitor, with additional activity against trial of single-agent PTK 787/ZK 222584 (PTK/ZK), a novel, epidermal growth factor receptor tyrosine kinase. Clin Cancer oral angiogenesis inhibitor, in patients with recurrent Res 9: 1546-1556, 2003. glioblastoma multiforme (GBM). J Clin Oncol ASCO Annual 91 Sandström M, Johansson M, Bergström P, Bergenheim AT and Meeting Proc 22(Suppl): 1512, 2004. Henriksson R: Effects of the VEGFR inhibitor ZD6474 in 80 Reardon DA, Friedman H, Yung WKA, Brada M, Conrad C, combination with radiotherapy and temozolomide in an Provenzale J, Jackson E, Serajuddin H, Chen B and Laurent D: orthotopic glioma model. J Neurooncol 88: 1-9, 2008. A phase I/II trial of PTK787/ZK 222584 (PTK/ZK), a novel, 92 Wachsberger PR, Burd R, Cardi C, Thakur M, Daskalakis C, oral angiogenesis inhibitor, in combination with either Holash J, Yancopoulos GD and Dicker AP: VEGF Trap in temozolomide or lomustine for patients with recurrent combination with radiotherapy improves tumor control in u87 glioblastoma multiforme (GBM). J Clin Oncol ASCO Annual glioblastoma. Int J Radiat Oncol Biol Phys 67: 1526-1537, 2007. Meeting Proc 22(Suppl): 1513, 2004. 93 De Groot JF, Wen PY, Lamborn K, Chang S, Cloughesy TF, 81 Reardon DA, Egorin MJ, Desjardins A, Vredenburgh JJ, Beumer Chen AP, DeAngelis LM, Mehta MP, Gilbert MR, Yung WK JH, Lagattuta TF, Gururangan S, Herndon JE 2nd, Salvado AJ and Prados MD: Phase II single arm trial of aflibercept in and Friedman HS: Phase I pharmacokinetic study of the vascular patients with recurrent temozolomide-resistant glioblastoma: endothelial growth factor receptor tyrosine kinase inhibitor NABTC 0601. J Clin Oncol ASCO Annual Meeting Proc vatalanib (PTK787) plus imatinib and hydroxyurea for 26(Suppl): 2020, 2008. malignant glioma. Cancer 115: 2188-2198, 2009 94 Guertin DA and Sabatini DM. Defining the role of mTOR in 82 Batchelor T, Sorensen AG, Ancukiewicz MD, Duda G, Louis cancer: Cancer Cell 12: 9-22, 2007. DN, Plotkin SR, Ivy P, Wen PY, Loeffler JS and Jain R: A phase 95 Hu X, Pandolfi PP, Li Y, Koutcher JA, Rosenblum M and II trial of AZD2171 (cediranib), an oral pan-VEGF receptor Holland EC: mTOR promotes survival and astrocytic tyrosine kinase inhibitor, in patients with recurrent glioblastoma. characteristics induced by PTEN/AKT signaling in glioblastoma. J Clin Oncol ASCO Annual Meeting Proc 25(Suppl): 2001, 2007. Neoplasia 7: 356-368, 2007. 83 Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, 96 Wang SI, Puc J, Li J Bruce JN, Cairns P, Sidransky D and Bello C, Deprimo S, Brega N, Massimini G, Armand JP, Scigalla Parsons R: Somatic mutations of PTEN in glioblastoma P and Raymond E: Safety, pharmacokinetic, and antitumor multiforme. Cancer Res 57: 4183-4186, 1997. activity of SU11248, a novel oral multitarget tyrosine kinase 97 Zhou XP, Li YJ, Hoang-Xuan K, Laurent-Puig P, Mokhtari K, inhibitor, in patients with cancer. J Clin Oncol 24: 25-35, 2006. Longy M, Sanson M, Delattre JY, Thomas G and Hamelin R: 84 Motzer RJ and Bukowski RM: Targeted therapy for metastatic Mutational analysis of the PTEN gene in gliomas: molecular renal cell carcinoma. J Clin Oncol 24: 5601-5608, 2006. and pathological correlations. Int J Cancer 84: 150-154, 1999. 85 Faivre S, Demetri G, Sargent W and Raymond E: Molecular 98 Sano T, Lin H, Chen X, Langford LA, Koul D, Bondy ML, basis for sunitinib efficacy and future clinical development. Nat Hess KR, Myers JN, Hong YK, Yung WK and Steck PA: Rev Drug Discov 6: 734-745, 2007. Differential expression of MMAC/PTEN in glioblastoma 86 Gollob JA, Rathmell WK, Richmond TM, Marino CB, Miller multiforme: relationship to localization and prognosis. Cancer EK, Grigson G, Watkins C, Gu L, Peterson BL and Wright JJ: Res 59: 1820-1824, 1999. Phase II trial of sorafenib plus interferon α-2b as first- or 99 Ermoian RP, Furniss CS, Lamborn KR, Basila D, Berger MS, second-line therapy in patients with metastatic renal cell cancer. Gottschalk AR, Nicholas MK, Stokoe D and Haas-Kogan DA: J Clin Oncol 25: 3288-3295, 2007. Dysregulation of PTEN and protein kinase B is associated with 87 Ryan CW, Goldman BH, Lara PN Jr, Mack PC, Beer TM, glioma histology and patient survival. Clin Cancer Res 8: 1100- Tangen CM, Lemmon D, Pan CX, Drabkin HA, Crawford ED; 1106, 2002. Southwest Oncology Group: Sorafenib with interferon α-2b as 100 Schmelzle T and Hall MN: TOR, a central controller of cell first-line treatment of advanced renal carcinoma: a phase II growth. Cell 103: 253-262, 2000. study of the Southwest Oncology Group. J Clin Oncol 25: 101 Castedo M, Ferri KF and Kroemer G: Mammalian target of 3296-3301, 2007. rapamycin (mTOR): pro- and anti-apoptotic. Cell Death Differ 88 Escudier B, Szczylik C, Hutson TE, Demkow T, Staehler M, 9: 99-100, 2002. Rolland F, Negrier S, Laferriere N, Scheuring UJ, Cella D, Shah S 102 Podsypanina K, Lee RT, Politis C, Hennessy I, Crane A, Puc J, and Bukowski RM: Randomized phase II trial of first-line Neshat M, Wang H, Yang L, Gibbons J, Frost P, Dreisbach V, treatment with sorafenib versus interferon α-2a in patients with Blenis J, Gaciong Z, Fisher P, Sawyers C, Hedrick-Ellenson L metastatic renal cell carcinoma. J Clin Oncol 27: 1280-1289, 2009. and Parsons R: An inhibitor of mTOR reduces neoplasia and 89 Wedge SR, Ogilvie DJ, Dukes M, Kendrew J, Chester R, normalizes p70/S6 kinase activity in Pten +/- mice. Proc Natl Jackson JA, Boffey SJ, Valentine PJ, Curwen JO, Musgrove Acad Sci USA 98: 10320-10325, 2001.

5183 ANTICANCER RESEARCH 29: 5171-5184 (2009)

103 Choe G, Horvath S, Cloughesy TF, Crosby K, Seligson D, Palotie 113 Moyal EC, Laprie A, Delannes M, Poublanc M, Catalaa I, A, Inge L, Smith BL, Sawyers CL and Mischel PS: Analysis of Dalenc F, Berchery D, Sabatier J, Bousquet P, De Porre P, the phosphatidylinositol 3’-kinase signaling pathway in Alaux B and Toulas C: Phase I trial of tipifarnib (R115777) glioblastoma patients in vivo. Cancer Res 63: 2742-2746, 2003. concurrent with radiotherapy in patients with glioblastoma 104 Rajasekhar VK, Viale A, Socci ND, Wiedmann M, Hu X and multiforme. Int J Radiat Oncol Biol Phys 68: 1396-1401, 2007. Holland EC: Oncogenic Ras and Akt signaling contribute to 114 Gilbert MR, Gaupp P, Liu V, Conrad C, Colman H, Groves M, glioblastoma formation by differential recruitment of existing Puduvalli V, Levin V, Hsu S, Horowitz J and Yung W: A phase mRNAs to polysomes. Mol Cell 12: 889-901, 2003. I study of temozolomide (TMZ) and the farnesyltransferase 105 Chang SM, Wen P, Cloughesy T, Greenberg H, Schiff D, Conrad inhibitor (FTI), lonafarnib (Sarazar, SCH66336) in recurrent C, Fink K, Robins HI, De Angelis L, Raizer J, Hess K, Aldape glioblastoma. J Clin Oncol ASCO Annual Meeting Proc K, Lamborn KR, Kuhn J, Dancey J and Prados MD; North 24(Suppl): 1556, 2006. American Brain Tumor Consortium and the National Cancer 115 Stupp R and Ruegg C. Integrin inhibitors reaching the clinic. Institute: Phase II study of CCI-779 in patients with recurrent J Clin Oncol 25: 1637-1638, 2007. glioblastoma multiforme. Invest New Drugs 23: 357-361, 2005. 116 Rüegg C, Postigo AA, Sikorski EE, Butcher EC, Pytela R and 106 Galanis E, Buckner JC, Maurer MJ, Kreisberg JI, Ballman K, Erle DJ: Role of integrin alpha 4 beta 7/alpha 4 beta P in Boni J, Peralba JM, Jenkins RB, Dakhil SR, Morton RF, lymphocyte adherence to fibronectin and VCAM-1 and in Jaeckle KA, Scheithauer BW, Dancey J, Hidalgo M, Walsh DJ; homotypic cell clustering. J Cell Biol 117: 179-189, 1992. North Central Cancer Treatment Group: Phase II trial of 117 Reardon DA, Nabors LB, Stupp R and Mikkelsen T. temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a Cilengitide: an integrin-targeting arginine-glycine-aspartic acid North Central Cancer Treatment Group Study. J Clin Oncol 23: peptide with promising activity for glioblastoma multiforme. 5294-5304, 2005. Expert Opin Investig Drugs 17: 1225-1235, 2008. 107 Rao RD, Mladek AC, Lamont JD, Goble JM, Erlichman C, 118 Nabors LB, Mikkelsen T, Rosenfeld SS, Hochberg F, Akella James CD and Sarkaria JN: Disruption of parallel and NS, Fisher JD, Cloud GA, Zhang Y, Carson K, Wittemer SM, converging signaling pathways contributes to the synergistic Colevas AD and Grossman SA: Phase I and correlative biology antitumor effects of simultaneous mTOR and EGFR inhibition study of cilengitide in patients with recurrent malignant glioma. in GBM cells. Neoplasia 7: 921-929, 2005. J Clin Oncol 25: 1651-1657, 2007. 108 Nguyen TD, Lassman AB, Lis E, Rosen N, Shaffer DR, Scher 119 Reardon DA, Fink KL, Mikkelsen T, Hochberg F, Akella NS, HI, Deangelis LM and Abrey LE: A pilot study to assess the Fisher JD, Cloud GA, Zhang Y, Carson K, Wittemer SM, tolerability and efficacy of RAD-001 (everolimus) with gefitinib Colevas AD and Grossman SA: Randomized phase II study of in patients with recurrent glioblastoma multiforme (GBM). J cilengitide, an integrin-targeting arginine-glycine-aspartic acid Clin Oncol ASCO Annual Meeting Proc 24(Suppl): 1507, 2006. peptide, in recurrent glioblastoma multiforme. J Clin Oncol 26: 109 Reardon DA, Quinn JA, Vredenburgh JJ, Gururangan S, 5610-5617, 2008. Friedman AH, Desjardins A, Sathornsumetee S, Herndon JE 120 Stupp R, Goldbrunner R, Neyns B, Schlegel U, Clement P, 2nd, Dowell JM, McLendon RE, Provenzale JM, Sampson JH, Grabenbauer GG, Hegi ME, Nippgen J, Picard M and Weller Smith RP, Swaisland AJ, Ochs JS, Lyons P, Tourt-Uhlig S, M: Phase I/IIa trial of cilengitide (EMD121974) and Bigner DD, Friedman HS and Rich JN: Phase 1 trial of gefitinib temozolomide with concomitant radiotherapy, followed by plus sirolimus in adults with recurrent malignant glioma. Clin temozolomide and cilengitide maintenance therapy in patients Cancer Res 12: 860-868, 2006. (pts) with newly diagnosed glioblastoma (GBM). J Clin Oncol 110 Fine HA, Puduvalli VK, Chamberlain MC, Carpentier AF, Cher ASCO Annual Meeting Proc 25(Suppl): 2000, 2007. L, Mason WP, van den Bent MJ, Hong S, Thornton D, Wick W. Enzastaurin (ENZ) versus lomustine (CCNU) in the treatment of recurrent, intracranial glioblastoma multiforme (GBM): A phase III study. J Clin Oncol ASCO Annual Meeting Proc 26(Suppl): 2005, 2008. 111 Tabatabai G, Frank B, Wick A, Lemke D, von Kürthy G, Obermüller U, Heckl S, Christ G, Weller M and Wick W: Synergistic antiglioma activity of radiotherapy and enzastaurin. Ann Neurol 61: 153-161, 2007. 112 Cloughesy TF, Wen PY, Robins HI, Chang SM, Groves MD, Fink KL, Junck L, Schiff D, Abrey L, Gilbert MR, Lieberman F, Kuhn J, DeAngelis LM, Mehta M, Raizer JJ, Yung WK, Aldape K, Wright J, Lamborn KR and Prados MD: Phase II trial of tipifarnib in patients with recurrent malignant glioma either receiving or not receiving enzyme-inducing antiepileptic Received July 27, 2009 drugs: a North American Brain Tumor Consortium Study. J Clin Revised November 9, 2009 Oncol 24: 3651-3656, 2006. Accepted November 11, 2009

5184