702
Views
1
CrossRef citations to date
0
Altmetric
Theme: Nervous system neoplasms - Meeting Report

Neuro-oncology: a selected review of ASCO 2011 abstracts

Pages 1371-1377 | Published online: 09 Jan 2014

Abstract

American Society of Clinical Oncology (ASCO), the largest clinical oncology meeting in the USA, meets annually and consistently provides an exciting forum to present new cancer clinical trials and research data. The ASCO 2011 neuro-oncology session, comprising of 3 days of presentations and over 100 abstracts, provided an overview of neuro-oncology, including both metastatic diseases of the CNS and primary brain tumors. This brief article attempts to highlight select abstracts presented at this years meeting in an organizational manner that will hopefully provide a portrait of the large and multifaceted meeting.

Metastatic disease of the CNS

Parenchymal brain metastases

Three abstracts were presented, two regarding the use of the biodegradable carmustine wafer, Gliadel®, implanted at the time of brain metastases (BM) resection and one evaluating the combined use of erlotinib, a tyrosine kinase EGF receptor (EGFR) inhibitor, with whole-brain irradiation (WBI) for BM resulting from non-small-cell lung cancer Citation[1–3]. A prospective single-arm Phase II trial of one to three BM treated with surgical resection, Gliadel implantation, stereotactic radiotherapy (RT) and deferred WBI evaluated neurocognitive outcome in 59 patients Citation[1]. The results suggested improvement in all three cognitive domains (memory, executive function and fine motor) following treatment as compared with baseline. In a retrospective study of 49 patients with BM treated with surgical resection, Gliadel implantation and WBI, local control was improved compared with an institutional historical control cohort treated with surgery and WBI only Citation[2].

A study of 40 patients with BM and non-small-cell lung cancer using erlotinib and WBI followed by erlotinib only suggested an improved overall survival (OS) relative to historical controls (10.9 vs 3.9 months) Citation[3].

Leptomeningeal disease

Five abstracts of special note were presented, three concerned with diagnosis and two with treatment of leptomeningeal disease (LMD) Citation[4–8]. In a study of 80 women with breast cancer and LMD, cerebrospinal fluid (CSF) CA15-3 was found diagnostically useful; however, the relationship to CSF cytology or MRI was not presented, making a determination of the utility of CSF CA15-3 difficult Citation[4]. A retrospective study of 21 patients suggested that CSF lactate, assessed by ventricular magnetic resonance spectroscopy, may be both sensitive and specific for LMD. However, whether CSF lactate determinations are useful compared with magnetic resonance spectroscopy obtained lactate levels was not mentioned in the abstract Citation[5]. Another retrospective study suggested that CSF cytology is most useful (as determined by the likelihood ratio) in patients with cancer (n = 81) and neurological findings compatible with LMD, as contrasted with patients without cancer (n = 403) or patients with cancer and no neurological symptoms (n = 41) Citation[6]. A retrospective study of 120 patients with LMD treated with intra-CSF liposomal cytarabine (80 by intraventricular and 40 by intralumbar administration) demonstrated a 14% incidence of permanent treatment-related neurological deficits, probably reflecting the long half-life of the chemotherapy and suggesting caution when using this agent Citation[7]. Last, a large retrospective study of 96 women with breast cancer-related LMD demonstrated that median survival was 3.5 months and survival was improved in women with good performance status, those who were HER2/neu negative or did not have triple negative breast cancer, and those who received timely LMD-directed therapy – that is, intra-CSF and systemic chemotherapy and RT Citation[8].

Primary brain tumors

Meningioma

Two abstracts were presented regarding the treatment of surgery and radiation refractory recurrent meningioma with SOM230 (pasireotide; n = 26; 17 with high-grade meningiomas) or PTK787 (vatalanib; n = 21; all with high-grade meningiomas). Both studies utilized targeted therapies; SOM230 is a somatostatin receptor agonist and PTK787 is a multifunctional tyrosine kinase inhibitor with anti-VEGFR2 activity Citation[9,10]. The overall radiographic response was meager (0% with SOM230; 5.8% with PTK787), although median progression-free survival (mPFS) was similar in both (˜4 months). PTK787 had a superior 6-month progression-free survival (PFS-6) of 37.5 versus 20% for SOM230. These studies represent an evolving trend in neuro-oncology for using novel molecular targeted therapies for primary brain tumors.

Medulloblastoma

All three articles presented related to treatment, two regarding upfront treatment and one at recurrence Citation[11–13]. Head Start III, an adjuvant dose-intense chemotherapy regimen (vincristine, cisplatin, cyclophosphamide, etoposide and high-dose methotrexate) with high-dose conditioning chemotherapy (carboplatin, etoposide and thiotepa) and peripheral stem cell transplantation, was utilized in a multinational multi-institutional study of 92 children Citation[11]. RT was deferred in all but patients over the age of 6 years with incomplete responses following adjuvant chemotherapy. Event-free survival at 3 years (47%) surpassed that seen in prior studies, including similar Head Start I and II trials, suggesting the feasibility of this approach for childhood medulloblastoma and the need for a randomized trial comparing it to standard infant/childhood adjuvant treatment regimens. Another small study evaluated an attenuated chemotherapy regimen for postcraniospinal irradiation treatment that utilized nine alternating cycles of three noncross resistant chemotherapies (vincristine, temozolomide and etoposide; vincristine and cyclophosphamide; and vincristine and carboplatin) Citation[12]. The utility of this study is to be examined in a larger randomized trial. Last, a small study (n = 47) suggested that recurrent medulloblastoma in a contemporary series is frequently metastatic (86% of patients in this small series) and more common than historically appreciated, presents on average 18 months after initial surgery and diagnosis, has a median OS of 6.8 months and a 3-year survival of 19% Citation[13].

Primary CNS lymphoma

Three articles were derived from the large (n = 411) German primary CNS lymphoma randomized trial of high-dose methotrexate (HD-MTX) with or without ifosfamide followed in patients with a complete response to observation or whole brain RT Citation[14–17]. The trial demonstrated no advantage to upfront RT and was further analyzed with respect to prognostic variables affecting survival, as well as chemotherapy-related toxicity Citation[18]. Prognosis was primarily a function of age (< or > 60 years) and Karnofsky performance status (< or > 70) and secondarily of gender (female OS > male OS) and BMI Citation[14]. These results recapitulate prior published data that was derived from a retrospective review of a large database Citation[15]. Similarly, chemotherapy-related toxicity was a function of low performance (<70), advanced age (>60 years), receipt of HD-MTX and ifosfamide (compared with HD-MTX only), elevated serum LDH and pre-existing pulmonary or cardiovascular disease Citation[16]. The German primary CNS lymphoma study also analyzed whether evidence of CSF dissemination at diagnosis affected outcome as determined by CSF cytology, polymerase chain reaction (PCR) gene rearrangement or MRI Citation[17]. Despite no intra-CSF chemotherapy being administered in this study, outcome in the 19% of patients with CSF dissemination was similar to that of patients without evidence of CSF dissemination. Two articles concerned recurrent disease and treatment Citation[19,20]. In a single institution study (n = 38), re-challenge with HD-MTX in patients previously responding to HD-MTX re-responded with a mPFS of 12 months and an overall radiologic response of 84% Citation[19]. In a small (n = 16) Adult Brain Tumor Consortium trial, the combination of temozolomide (TMZ) and rituximab proved ineffective with an mPFS of 7 weeks Citation[20].

Gliomas

Low-grade gliomas

Two studies were notable, one evaluating the effect of RT treatment on seizure control and the other identifying a new low-grade glioma (LGG) entity, the so-called triple-negative LGG Citation[21,22]. RT, administered to nearly all patients with LGG, has been suggested as having salutatory benefits with regard to control of tumor-related epilepsy. In a small study of 40 patients with pretreatment epilepsy, nearly 70% improved following the administration of RT, suggesting that seizure control (defined as a greater than 50% reduction in seizures) may indeed be achieved by RT Citation[21]. Metellus et al. define triple negative LGG as gliomas without 1p19q codeletion and possessing wild-type p53 and IDH1/2, a molecular signature that predicts poor outcome and is more commonly seen in older patients with LGG and large infiltrative tumors of the insula Citation[23]. Molecular stratification of LGG has not yet entered clinical trials, but will probably be relevant with future studies.

Anaplastic gliomas

A newly diagnosed trial, the Temozolomide and BCNU for Newly Diagnosed Anaplastic Oligodendroglioma (TEMOBIC) study by the French Association des Neuro-Oncologues d’Expression Française consortium evaluated 54 patients with anaplastic oligodendroglial tumors treated with the combination of TMZ and carmustine (BCNU) for six cycles followed by consolidative RT Citation[24]. Overall radiologic response for chemotherapy was 38%, mPFS was 15.4 months and median OS (mOS) 25 months. Grade 3 or higher heme-related toxicity was 50 and 6% of patients sustained a treatment-related death. The utility of this approach is uncertain given the large randomized trials that have recently opened for newly diagnosed anaplastic gliomas (AG) – that is, the Uni- or Non-1p19q Deleted Trial for Newly Diagnosed Anaplastic Glioma (CATNON; RT with or without TMZ followed by observation or TMZ) for non- or uni-deleted AG and the 1p19q Codeletion Trial for Newly Diagnosed Anaplastic Gliomas (CODEL; randomization to TMZ vs RT vs TMZ plus RT) for codeleted AG. A retrospective review of 96 elderly (aged >65 years) patients with AG suggests poor survival in patients with anaplastic astrocytoma (mOS: 5.4 months), as well as in patients with compromised performance status (Karnofsky <70) Citation[25].

Glioblastoma

Upfront trials

outlines the 15 clinical trials presented regarding the treatment of newly diagnosed glioblastoma (GBM) Citation[24–40]. The most anticipated trial, RTOG 0525, was a randomized trial of 833 patients comparing post-RT standard dose TMZ (sdTMZ; standard of care [SOC]) to dose-dense TMZ (ddTMZ) Citation[24]. No statistical difference was seen between these arms with respect to mPFS (5.5 vs 6.6 months), mOS (18.9 vs 16.8 months) or in methylation status. In addition, there was increased toxicity in the ddTMZ arm (grade 3+: 27 vs 19%). The study did confirm in a prospective manner the predictive value of methylguanine methyltransferase (MGMT) methylation in newly diagnosed GBM. Two other randomized Phase III trials were presented, the nimotuzumb (an EGFR monoclonal antibody) trial and the Avastin for Newly Diagnosed Glioblastoma (AvaGlio) trial (upfront bevacizumab in conjunction with SOC), which recently completed accrual, but are without outcome results Citation[25,26]. The nimotuzumab trial compared standard therapy (RT plus TMZ followed by TMZ) with or without nimotuzumb in 150 patients Citation[25]. No survival advantage was seen, although in a subset analysis of tumors with amplified EGFR (˜20% of all GBM), a suggestion of improved outcome was reported (14.9 vs 19.4 months). Eight trials (nine including the AvaGlio trial) were presented; however, all represent work in progress and consequently had no outcome results Citation[26–29,31,32,34–36]. The majority of these trials are single institution and single arm utilizing upfront bevacizumab or cilengitide (an integrin inhibitor) with SOC. A novel trial design treated 57 patients with unmethylated MGMT and utilized enzastaurin (a protein kinase C inhibitor) in conjunction with RT followed by enzastaurin Citation[30]. This represents the first completed trial of newly diagnosed patients with GBM selected for MGMT status and deferring TMZ use based on prior studies, suggesting little benefit from TMZ in this patient cohort Citation[41]. Using PFS-6 as the primary end point, the study failed to achieve the primary objective. Two other small trials addressed novel approaches to newly diagnosed GBM Citation[37,38]. In one, a radiation sensitizer paclitaxel polyglutamate polymer was utilized during TMZ-based chemoradiotherapy (CRT) and judged to sensitize the tumor based upon the high incidence of apparent pseudoprogression Citation[37]. In the second trial, a dendritic cell vaccine was used post-RT and generated by the use of six tumor-associated antigens. mPFS in this highly selected cohort was 16 months and as a result is being taken forward as a randomized Phase II study Citation[38]. Three population-based studies were presented Citation[42–44]. In a Canadian study of 433 patients, only 44% received the SOC (CRT) for newly diagnosed GBM Citation[42]. An analysis of the Surveillance, Epidemiology and End Results database of 7022 patients revealed an improvement in mOS and 2-year OS over three epochs (2000–2001, 2002–2003 and 2005–2006) suggesting continued improvement in care over time with the introduction of CRT Citation[43]. In an Austrian database, a similar improvement of OS was seen in the contemporary cohort treated with CRT (n = 375) compared with a historical control treated primarily with RT only Citation[44].

Salvage trials

outlines the 12 clinical trials presented regarding the treatment of recurrent GBM Citation[45–56]. In the only randomized trial, afatinib, an irreversible tyrosine kinase EGFR inhibitor, was compared with ddTMZ and to the combination of ddTMZ and afatinib in 120 patients with first recurrent GBM Citation[45]. Afatinib was found not to be active, whereas ddTMZ with or without afatinib demonstrated a PFS-6 of approximately 20%, a result not dissimilar to the Canadian TMZ re-challenge RESCUE trial Citation[57]. In an analysis of patients with overexpressed variant 3 EGFR, a suggestion of afatinib response was observed. In a small study of 48 patients with recurrent GBM, ddTMZ was utilized and demonstrated similar efficacy to that seen in the afatinib trial (mPFS: 10 weeks; PFS-6: 23%) Citation[49]. A Phase I study evaluated the novel nitrosourea, laromustine, given in combination with ddTMZ and determined a maximum tolerated dose of laromustine (100 mg/m2 every 4 weeks) in combination with ddTMZ Citation[56]. Five articles discussed the continuing use of bevacizumab for recurrent GBM Citation[46,47,50,53,55]. In three trials of these articles, a novel partner (fotemustine, a nitrosourea; bortezomib, a proteasomal inhibitor; or panobinostat, a histone deacetylase inhibitor) was combined with bevacizumab Citation[46,50,55]. Results appear not dissimilar to single agent bevacizumab; however, increased toxicity was observed. Another study combined bevacizumab with stereotactic RT, suggesting both feasibility and efficacy not dissimilar to a prior published article Citation[53,58]. In a large study of 95 patients treated following failure on bevacizumab, outcome was compared in two subgroups. In one subgroup (53 patients), treatment entailed the continuation of bevacizumab with another agent, while in the comparator group (42 patients) treatment with bevacizumab was discontinued and another treatment administered. mOS was 6.1 months in the bevacizumab continuation group compared with 4.5 months in the bevacizumab discontinuation group, suggesting a marginal advantage to continuing bevacizumab. Four articles utilized novel agents to treat bevacizumab-naive patients with recurrent GBM Citation[48,51,52,54]. A combination of sorafenib (a multifunctional tyrosine kinase inhibitor that has anti-VEGF receptor activity) and temsirolimus (an mTOR inhibitor) was found to be inactive (mPFS: 2.6 months; mOS 4.2 months) and toxic in 37 patients Citation[48]. Similarly, a study of 36 patients with sorafenib and metronomic (low dose daily) TMZ demonstrated limited activity, similar to the aforementioned trial of sorafenib and temsirolimus Citation[52]. A study of 31 patients with the novel mitotic spindle and vascular disrupting agent verubilin was found to be inactive (mPFS: 1.8 months). Last, an interesting trial of the novel nitrosourea fotemustine in 119 patients suggested similar activity to that of the Canadian TMZ RESCUE trial wherein three groups of patients were analyzed with respect to failure on sdTMZ (progression on sdTMZ and within 6 months of completion of CRT; progression on sdTMZ while on post-RT sdTMZ for >6 months and progression after completion of post-RT sdTMZ) Citation[51,57]. These data confirm the nitrosourea to be a valid salvage therapy for GBM progression following sdTMZ similar to that reported in the enzastaurin and cediranib trials Citation[28,59].

Molecular correlatives

Five articles were presented in relation to molecular correlatives, four concerning GBM and one AG Citation[60–64]. In an analysis of the German Glioma Network database, 233 elderly patients (median age: 74 years) underwent MGMT promoter methylation determination by two methods: DNA pyrosequencing and methylation-specific polymerase chain reaction (MSP-PCR) Citation[61]. Interestingly, MGMT promoter methylation was found to be more common in the elderly (58%) and, as seen in other adult cohorts, had improved survival when treated with TMZ only or CRT. No benefit was seen with the administration of TMZ to the unmethylated MGMT elderly patients. Last, MGMT methylation appeared predictive of response to TMZ in the elderly. In a study of 418 patients with GBM treated primarily with CRT, a combined analysis of MGMT was assessed using immunohistochemistry (IHC; <30%>), a measure of protein expression and promoter methylation by both MSP-PCR (9 CpG sites interrogated) and bisulfite sequencing (BiSeq; 24 CpG sites investigated) Citation[63]. All three determinations were predictive individually; however, BiSeq was superior to MSP-PCR, which was superior to IHC. The combination was most predictive and outcome of patients with low IHC expression was a function of promoter methylation status. A small study of 15 patients with paired tumor samples obtained at initial surgery and at time of recurrence analyzed for MGMT methylation status demonstrated concordance for the majority of patients (>85%) Citation[64]. In an analysis of the RTOG 0525 trial, four molecular biomarkers were defined in 82% of all submitted tumors: MSP-PCR, glioma CpG island methylation phenotype, a microarray mRNA-based panel of 19 genes and IDH1/2 mutational status Citation[60]. Using this biomarker panel, four molecular classes were defined, each with a distinct mOS ranging from 12 to 26 months. This aggregate biomarker panel may prove useful for future stratification of newly diagnosed GBM. Last, in a small study of 43 anaplastic oligodendroglial tumors, isocitrate dehydrogenase 1 or 2 (IDH1/2) mutations were found in 56% of tumors and were associated with improved PFS and OS relative to wild-type IDH1/2 tumors, suggesting that IDH1/2 mutational status is a prognostic molecular signature in AG Citation[62].

Table 1. Up-front glioblastoma trials.

Table 2. Recurrent glioblastoma trials.

Financial & competing interests disclosure

The author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Ewend MG, Meyers CA, Silva E, Booth-Jones M, Jain S, Brem S. Effects of surgery with BCNU wafer placement on neurocognitive function in patients with one to three brain metastases. J. Clin. Oncol.29(15 Suppl., part I of II), 143s (2011) (Abstract 2015).
  • Brem S, Sampath R, Yu D, Staller A, Obadia M, Ewend MG. Stereotactic volumetric resection, intracavitary carmustine wafers, and radiation therapy for brain metastases: the Moffitt Cancer Center experience. J. Clin. Oncol.29(15 Suppl., part I of II), 159s (2011) (Abstract 2077).
  • Welsh J, Amini A, Kim ES et al. Phase II study of erlotinib with concurrent whole-brain radiation therapy for patients with brain metastases from non-small lung cancer. J. Clin. Oncol.29(15 Suppl., part I of II), 147s (2011) (Abstract 2031).
  • Salinque S, Griot M, Rodrigues S et al. Value of CA 15-3 detection in cerebrospinal fluid (CSF) of breast cancer leptomeningeal metastases. J. Clin. Oncol.29(15 Suppl., part I of II), 158s (2011) (Abstract 2075).
  • Weston CL, Zalatimo O, Sheehan J et al. Novel use of MR spectroscopy in detecting CSF lactate for diagnosis of neoplastic meningitis. J. Clin. Oncol.29(15 Suppl., part I of II), 157s (2011) (Abstract 2071).
  • Zoccoli CM, Zalatimo O, Glantz MJ. Is cerebrospinal fluid cytology a useful test? J. Clin. Oncol.29(15 Suppl., part I of II), 159s (2011) (Abstract 2079).
  • Chamberlain MC, Johnston SK. Neurotoxicity of intra-CSF liposomal cytarabine in the treatment of leptomeningeal metastases. J. Clin. Oncol.29(15 Suppl., part I of II), 155s (2011) (Abstract 2062).
  • Le Rhun E, Kotecki N, Zairi F et al. Prospective follow-up of 96 patients with breast cancer with leptomeningeal metastasis recruited from 2007 to 2010. J. Clin. Oncol.29(15 Suppl., part I of II), 162s (2011) (Abstract 2089).
  • Norden AD, Hammond S, Drappatz J et al. Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma. J. Clin. Oncol.29(15 Suppl., part I of II), 150s (2011) (Abstract 2040).
  • Grimm SA, Chamberlain MC, Chandler J et al. A Phase II trial PTK787/ZK 222584 (PTK787) in recurrent high-grade meningioma. J. Clin Oncol.29(15 Suppl., part I of II), 151s (2011) (Abstract 2046).
  • Dhall G, Ji L, Haley K et al. Outcome of infants and young children with newly diagnosed medulloblastoma treated on Head Start III protocol. J. Clin. Oncol.29(15 Suppl., part I of II), 142s (2011) (Abstract 2011).
  • Dagri JN, Evans A, Torkildson J et al. Feasibility pilot of attenuated maintenance chemotherapy for adolescents and adults with newly diagnosed localized medulloblastoma and other primitive neuroectodermal tumors. J. Clin. Oncol.29(15 Suppl., part I of II), 160s (2011) (Abstract 2081).
  • Koschmann C, Schmidt KL, Geyer JR, Leary S. Survival after recurrence of medulloblastoma in the contemporary era. J. Clin Oncol.29(15 Suppl., part I of II), 157s (2011) (Abstract 2068).
  • Jahnke K, Korfel A, Martus P et al. Prognostic factors for response and survival in primary central nervous system lymphoma (PCNSL) from a randomized Phase III trial (G-PCNSL-SG1). J. Clin. Oncol.29(15 Suppl., part I of II), 141s (2011) (Abstract 2004).
  • Abrey LE, Ben-Porat L, Panageas KS et al. Primary central nervous system lymphoma: the Memorial Sloan-Kettering Cancer Center prognostic model. J. Clin. Oncol.24(36), 5711–5715 (2006).
  • Martus P, Jahnke K, Korfel A et al. Prognostic factors for chemotherapy-related toxicity in primary central nervous system lymphoma (PCNSL) treated with high-dose methotrexate (HDMTX) with or without ifosfamide: results from a German Phase III trial (G-PCNSL-SG-1). J. Clin. Oncol.29(15 Suppl., part I of II), 141s (2011) (Abstract 2005).
  • Korfel A, Fischer L, Martus P et al. Impact of meningeal dissemination (MD) on outcome in primary CNS lymphoma in the G-PCNSL-SG1 trial. J. Clin. Oncol.29(15 Suppl., part I of II), 146s (2011) (Abstract 2026).
  • Thiel E, Korfel A, Martus P et al. High-dose methotrexate with or without whole brain radiotherapy for primary CNS lymphoma (G-PCNSL-SG-1): a Phase 3, randomized non-inferiority trial. Lancet Oncology11(11), 1036–1047 (2010).
  • Pentsova E, DeAngelis LM, Omuro AM. Methotrexate (MTX) rechallenge for recurrent primary CNS lymphoma (PCNSL). J. Clin. Oncol.29(15 Suppl., part I of II), 146s (2011) (Abstract 2025).
  • Nayak L, Abrey LE, Drappatz J et al. Multicenter Phase II trial of temozolomide (TMZ) and rituximab (RIT) for recurrent primary CNS lymphoma (PCNSL): North American Brain Tumor Consortium (NABTC) study 05–01. J. Clin. Oncol.29(15 Suppl., part I of II), 149s (2011) (Abstract 2039).
  • Chinot OL, Hoang-Xuan K, Fabbro M et al. TEMOBIC: a ANOCEF Phase II study of BCNU and temozolomide (TMZ) combination prior to radiotherapy (RT) in anaplastic oligodengroglial gliomas (AOG). J. Clin. Oncol.29(15 Suppl., part I of II), 148s (2011) (Abstract 2034).
  • Satra A, Hashemi-Sadraei N, Bawa HS et al. Prognostic factors in elderly patients with grade 3 gliomas. J. Clin. Oncol.29(15 Suppl., part I of II), 163s (2011) (Abstract 2093).
  • Metellus P, Coulibaly B, Colin C et al. Triple negative, low grade gliomas: a highly aggressive tumor with dismal prognosis. J. Clin. Oncol.29(15 Suppl., part I of II), 145s (2011) (Abstract 2023).
  • Gilbert MR, Wang M, Aldape KD et al. RTOG 0525: a randomized Phase III trial comparing standard adjuvant temozolomide (TMZ) with a dose-dense (dd) schedule in newly diagnosed glioblastoma (GBM). J. Clin. Oncol.29(15 Suppl., part I of II), 141s (2011) (Abstract 2006).
  • Bach F, Westphal M; OSAG-101 Study Group. Current status of a Phase III trial of nimotuzumab (ti-EGF-R) in newly diagnosed glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 154s (2011) (Abstract 2059).
  • Chinot OL, Wick W, Saran F et al. AVAglio: a Phase III trial of bevacizumab added to standard radiotherapy and temozolomide in patients with newly diagnosed glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 16s (2011) (Abstract TPS136).
  • Verschaeve V, D’Hondt LA, Verbeke LM et al. CeCil: a randomized, noncomparative Phase II clinical trial of the effect of radiation therapy (RT) plus temozolomide (TMZ) combined with cilengitide or cetuximab on the 1-year overall survival of patients with newly diagnosed MGMT-promoter unmethylated glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 15s (2011) (Abstract TPS134).
  • Chauffert B, Feuvret L, Bonnetain F et al. Randomized multicenter Phase II trial of irinotecan and bevacizumab as neoadjuvant and adjuvant to temozolomide-based chemoradiation versus chemoradiation for unresectable glioblastoma: interim results of the TEMAVIR study from ANOCEF group. J. Clin. Oncol.29(15 Suppl., part I of II), 147s (2011) (Abstract 2029).
  • Hofland KF, Hansen S, Sorensen M et al. Randomized Phase II study of neoadjuvant bevacizumab and irinotecan versus bevacizumab and temozolomide followed by concomitant chemoreaadiotherapy in newly diagnosed primary glioblastoma multiforme. J. Clin. Oncol.29(15 Suppl., part I of II), 153s (2011) (Abstract 2052).
  • Wick W, Steinbach J, Combs SE et al. Enzastaurin (ENZ) before and concomitant with radiation therapy (RTX) followed by ENZ maintenance therapy in patients with newly diagnosed glioblastoma (GBM) without hypermethylation of the O6-methylguanyl DNA-methyltransferase (MGMT) promoter: a multicenter, open-label, uncontrolled Phase II study. J. Clin. Oncol.29(15 Suppl., part I of II), 141s (2011) (Abstract 2007).
  • Khasraw M, McCowatt S, Kerestes Z, Buyse ME, Back M, Wheeler H. Radiotherapy (RT), temozolomide (TMZ), procarbazine (PCB, and the integrin inhibitor cilengitide in patients with glioblastoma (GBM) without methylation of the MGMT gene promoter (ExCentric). J. Clin. Oncol.29(15 Suppl., part I of II), 15s (2011) (Abstract TPS133).
  • Omuro AMP, Beal K, Karimi S et al. Phase II study of bevacizumab (BEV), temozolomide (TMZ), and hypofractionated stereotactic radiotherapy (HFSRT) for newly diagnosed glioblastoma (GBM). J. Clin. Oncol.29(15 Suppl., part I of II), 147s (2011) (Abstract 2028).
  • Lou E, Reardon DA, Peters K et al. Upfront bevacizumab with temozolomide or with temozolomide and irinotecan for unresectable or multifocal glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 153s (2011) (Abstract 2055).
  • Quant EC, Batchelor T, Lassman AB et al. Preliminary results from a multicenter, phase II, randomized, noncomparative clinical trial of radiation and temozolomide with or without vandetanib in newly diagnosed glioblastoma (GBM). J. Clin. Oncol.29(15 Suppl., part I of II), 157s (2011) (Abstract 2069).
  • Beauchesnes PD, Faure G, Noel G et al. Concurrent, 3-times daily ultrafractionated radiation therapy and temozolomide for newly inoperable glioblastoma: TEMOFRAC a Phase II trial. J. Clin. Oncol.29(15 Suppl., part I of II), 158s (2011) (Abstract 2073).
  • Vredenburgh JJ, Desjardins A, Reardon DA et al. Bevacizumab, temozolomide, and radiation therapy followed by bevacizumab, temozolomide, and oral topotecan for newly-diagnosed glioblastoma multiforme (GBM). J. Clin. Oncol.29(15 Suppl., part I of II), 164s (2011) (Abstract 2098).
  • Jeyapalan SA, Goldmann M, Donahur J et al. A Phase II study of paclitaxel poliglumex (PPX), temozolomide (TMX), and radiation (RT) for newly diagnosed high-grade gliomas. J. Clin. Oncol.29(15 Suppl., part I of II), 149s (2011) (Abstract 2036).
  • Phuphanich S, Wheeler CJ, Rudnick J et al. Glioma-associated antigens associated with prolonged survival in a Phase I study of ICT-107 for patients with newly diagnosed glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 150s (2011) (Abstract 2042).
  • DeGroot JF, Cloughesy T, Lieberman FS et al. Phase I study of aflibercept (VEGF Trap) and temozolomide in newly diagnosed, high-grade glioma. J. Clin. Oncol.29(15 Suppl., part I of II), 150s (2011) (Abstract 2043).
  • Wen PY, Puduvalli VK, Kuhn JG et al. Phase I study of vorinostat in combination with temozolomide in patients with malignant gliomas. J. Clin. Oncol.29(15 Suppl., part I of II), 148s (2011) (Abstract 2032).
  • Hegi ME, Diserens AC, Gorlia T et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med.352(10), 997–1003 (2005).
  • Lwin Z, MacFadden D, AL-Zahrani A et al. A population-based study of glioblastom multiforme (GBM) in the new stupp paradigm: have we improved outcome? J. Clin. Oncol.29(15 Suppl., part I of II), 143s (2011) (Abstract 2012).
  • Koshy M, Villano JL, Dolecek TA et al. Improved survival time trends for glioblastoma using the SEER 17 population-based registries. J. Clin. Oncol.29(15 Suppl., part I of II), 143s (2011) (Abstract 2013).
  • Preusser M, Woehrer MA, Zielonke N et al. Real-life survival in unselected adult glioblastoma patients: a population-based study. J. Clin. Oncol.29(15 Suppl., part I of II), 143s (2011) (Abstract 2014).
  • Eisenstat DD, Nabors LB, Mason WP et al. A Phase II study of daily afatinib (BIBW 2992) with or without temozolomide (21/28 days) in the treatment of patients with recurrent glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 142s (2011) (Abstract 2010).
  • Soffietti R, Trevisan E, Ruda R et al. Phase II trial of bevacizumab with fotemustine in recurrent glioblastoma: final results of a multicenter study of AINO (Italian Association of Neuro-oncology). J. Clin. Oncol.29(15 Suppl., part I of II), 146s (2011) (Abstract 2027).
  • Reardon DA, Vredenburgh JJ, Desjardins A et al. Bevacizumab (BV) continuation following BV progression: meta-analysis of five consecutive recurrent glioblastoma (GBM) trials. J. Clin. Oncol.29(15 Suppl., part I of II), 147s (2011) (Abstract 2030).
  • Jaeckle KA, Schiff D, Anderson SK et al. NCCTG N0572 Phase I/II trial of sorafenib and temsirolimus in patients with recurrent glioblastoma: a North Central Cancer Treatment Group study. J. Clin. Oncol.29(15 Suppl., part I of II), 148s (2011) (Abstract 2033).
  • Hammond S, Norden AD, Lesser GJ et al. Phase II study of dose-intense temozolomide in recurrent glioblastoma: Phase II study of dose-intense temozolomide in recurrent glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 149s (2011) (Abstract 2038).
  • Bota DA, Erogluz, Reardon DA et al. Phase II clinical trial of bortezomib and bevacizumab combination in recurrent glioblastoma. J. Clin. Oncol.29(15 Suppl., part I of II), 154s (2011) (Abstract 2056).
  • Paccepelo ALolli I, Scocciantis et al. Efficacy of nitrosourea-based chemotherapy in recurrent malignant glioma according to time to adjuvant temozolomide failure: a pooled analysis. J. Clin. Oncol.29(15 Suppl., part I of II), 155s (2011) (Abstract 2060).
  • Zustovich F, Landi L, Lombaardi G et al. Sorafenib plus daily low dose temozolomide for relapsed glioblastoma: a Phase II study. J. Clin. Oncol.29(15 Suppl., part I of II), 160s (2011) (Abstract 2080).
  • Cuneo KC, Cabrera AR, Sampson JH et al. Safety results from prospective study of concurrent radiosurgery and bevacizumab for recurrent malignant glioma. J. Clin. Oncol.29(15 Suppl., part I of II), 160s (2011) (Abstract 2082).
  • Kim LJ, Chamberlain MC, Zhu J et al. Phase II study of verubulin (MPC-6827) for the treatment of subjects with recurrent glioblastoma naive to treatment with bevacizumab. J. Clin. Oncol.29(15 Suppl., part I of II), 162s (2011) (Abstract 2088).
  • Drappatz J, Raizer JJ, Schiff D et al. A Phase I trial of LBH589 and bevacizumab for recurrent high-grade glioma (HGG). J. Clin. Oncol.29(15 Suppl., part I of II), 152s (2011) (Abstract 2050).
  • Raizer JJ, Rice L, Rademaker A et al. A Phase I trial of laromustine (VPN40101M) and temozolomide for patients with malignant gliomas in first relapse of progression. J. Clin. Oncol.29(15 Suppl., part I of II), 156s (2011) (Abstract 2064).
  • Perry JR, Mason WP, Belanger K et al. The temozolomide RESCUE study: a Phase II trial of continuous (28/28) dose-intense temozolomide (TMZ) after progression on conventional 5/28 day TMZ in patients with recurrent malignant glioma. J. Clin. Oncol.26(15 Suppl.), (2010) (Abstract 2008).
  • Gutin PH, Iwamoto FM, Beal K et al. Safety and efficacy of bevacizumab with hypofractionated stereotactic irradiation for recurrent malignant gliomas. Int. J. Radiat. Oncol. Biol. Phys.75(1), 156–63 (2009).
  • Batchelor T, Mulholland P, Neyns B et al. A Phase III randomized study comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, with lomustine alone in recurrent glioblastoma patients. Neuro Oncol.12(Suppl 4), iv69 (2010) (Abstract OT-25).
  • Aldape KD, Wang M, Sulman EP et al. RTOG 0525: Molecular correlates from a randomized Phase III trial of newly diagnosed glioblastoma. J. Clin. Oncol.29(18S, part II of II), 779s (2011) (Abstract LBA2000).
  • Weller M, Hentschel B, Felsberg J et al. Impact of MGMT promoter methylation in glioblastoma of the elderly. J. Clin. Oncol.29(15 Suppl., part I of II), 140s (2011) (Abstract 2001).
  • Frenel J, Leux C, Loussouarn D et al. Predictive value of IDH1 mutation assessed by immunohistochemistry and DNA sequencing in WHO grade 3 oligodendrogliomas. J. Clin. Oncol.29(15 Suppl., part I of II), 140s (2011) (Abstract 2002).
  • Lai A, Lalezari S, Chou AP et al. Prediction of GBM outcome using combined analysis of MGMT protein expression and promoter methylation. J. Clin. Oncol.29(15 Suppl., part I of II), 140s (2011) (Abstract 2003).
  • Bertolini F, Baraldi C, Zunarelli E et al. O(6)-methylguanine DNA-methyltransferase (MGMT) in glioblastoma: analysis on first and following surgeries. J. Clin. Oncol.29(15 Suppl., part I of II), 163s (2011) (Abstract 2097).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.