Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
CNS Oncol ; 12(3): CNS102, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37462385

ABSTRACT

Aim: The EMulate Therapeutics Voyager™ is a simple, wearable, home-use device that uses an alternating electromagnetic field to alter biologic signaling within cells. Objective: To assess the safety/feasibility of the Voyager in the treatment of recurrent glioblastoma (rGBM). Methods: In this study, patients with rGBM were treated with Voyager as monotherapy or in combination with standard chemotherapy at the Investigator's discretion. Safety was assessed by incidence of adverse events associated with the Voyager. Patients were followed until death. Results: A total of 75 patients were enrolled and treated for at least one day with the Voyager (safety population). Device-related adverse events were uncommon and generally did not result in interruption or withdrawal from treatment. There were no serious adverse events associated with Voyager. A total of 60 patients were treated for at least one month (clinical utility population). The median progression-free survival (PFS) was 17 weeks (4.3 months) in the Voyager only group (n = 24) and 21 weeks (5.3 months) in the Voyager + concurrent therapy group (n = 36). The median overall survival (OS) was 7 months in the Voyager only group and 9 months in the Voyager + concurrent therapy group. In patients treated with Voyager + concurrent therapy, the median OS for patients enrolled with their 1st or 2nd recurrence (n = 26) was 10 months, while in patients enrolled with their 3rd or 4th recurrence (n = 10) OS was 7 months. Conclusion: The data support the safety and feasibility of the Voyager for the treatment of rGBM. Further prospective study of the device is warranted. Trial Registration Number: NCT02296580 (ClinicalTrials.gov).


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Brain Neoplasms/drug therapy , Feasibility Studies , Glioblastoma/drug therapy , Neoplasm Recurrence, Local , Prospective Studies
2.
J Natl Compr Canc Netw ; 21(1): 12-20, 2023 01.
Article in English | MEDLINE | ID: mdl-36634606

ABSTRACT

The NCCN Guidelines for Central Nervous System (CNS) Cancers focus on management of the following adult CNS cancers: glioma (WHO grade 1, WHO grade 2-3 oligodendroglioma [1p19q codeleted, IDH-mutant], WHO grade 2-4 IDH-mutant astrocytoma, WHO grade 4 glioblastoma), intracranial and spinal ependymomas, medulloblastoma, limited and extensive brain metastases, leptomeningeal metastases, non-AIDS-related primary CNS lymphomas, metastatic spine tumors, meningiomas, and primary spinal cord tumors. The information contained in the algorithms and principles of management sections in the NCCN Guidelines for CNS Cancers are designed to help clinicians navigate through the complex management of patients with CNS tumors. Several important principles guide surgical management and treatment with radiotherapy and systemic therapy for adults with brain tumors. The NCCN CNS Cancers Panel meets at least annually to review comments from reviewers within their institutions, examine relevant new data from publications and abstracts, and reevaluate and update their recommendations. These NCCN Guidelines Insights summarize the panel's most recent recommendations regarding molecular profiling of gliomas.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Adult , Humans , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/therapy , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Central Nervous System , Mutation
3.
Clin Cancer Res ; 28(4): 594-602, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34561269

ABSTRACT

Therapeutic advances for glioblastoma have been minimal over the past 2 decades. In light of the multitude of recent phase III trials that have failed to meet their primary endpoints following promising preclinical and early-phase programs, a Society for Neuro-Oncology Think Tank was held in November 2020 to prioritize areas for improvement in the conduct of glioblastoma clinical trials. Here, we review the literature, identify challenges related to clinical trial eligibility criteria and trial design in glioblastoma, and provide recommendations from the Think Tank. In addition, we provide a data-driven context with which to frame this discussion by analyzing key study design features of adult glioblastoma clinical trials listed on ClinicalTrials.gov as "recruiting" or "not yet recruiting" as of February 2021.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Glioblastoma/therapy , Humans , Research Design
4.
J Natl Compr Canc Netw ; 18(11): 1537-1570, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33152694

ABSTRACT

The NCCN Guidelines for Central Nervous System (CNS) Cancers focus on management of adult CNS cancers ranging from noninvasive and surgically curable pilocytic astrocytomas to metastatic brain disease. The involvement of an interdisciplinary team, including neurosurgeons, radiation therapists, oncologists, neurologists, and neuroradiologists, is a key factor in the appropriate management of CNS cancers. Integrated histopathologic and molecular characterization of brain tumors such as gliomas should be standard practice. This article describes NCCN Guidelines recommendations for WHO grade I, II, III, and IV gliomas. Treatment of brain metastases, the most common intracranial tumors in adults, is also described.


Subject(s)
Astrocytoma , Brain Neoplasms , Central Nervous System Neoplasms , Glioma , Adult , Astrocytoma/diagnosis , Astrocytoma/therapy , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Central Nervous System , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/therapy , Glioma/diagnosis , Glioma/therapy , Humans , Practice Guidelines as Topic
5.
J Neurooncol ; 137(3): 639-644, 2018 May.
Article in English | MEDLINE | ID: mdl-29332185

ABSTRACT

Gliomas are the most common type of malignant primary brain tumor and few risk factors have been linked to their development. Handedness has been associated with several pathologic neurological conditions such as schizophrenia, autism, and epilepsy, but few studies have evaluated a connection between handedness and risk of glioma. In this study, we examined the relationship between handedness and glioma risk in a large case-control study (1849 glioma cases and 1354 healthy controls) and a prospective cohort study (326,475 subjects with 375 incident gliomas). In the case-control study, we found a significant inverse association between left handedness and glioma risk, with left-handed persons exhibiting a 35% reduction in the risk of developing glioma [odds ratio (OR) = 0.65, 95% confidence interval (CI) 0.51-0.83] after adjustment for age, gender, race, education, and state of residence; similar inverse associations were observed for GBM (OR = 0.69, 95% CI 0.52-0.91), and non-GBM (OR = 0.59, 95% CI 0.42-0.82) subgroups. The association was consistent in both males and females, and across age strata, and was observed in both glioblastoma and in lower grade tumors. In the prospective cohort study, we found no association between handedness and glioma risk (hazards ratio = 0.92, 95% CI 0.67-1.28) adjusting for age, gender, and race. Further studies on this association may help to elucidate mechanisms of pathogenesis in glioma.


Subject(s)
Brain Neoplasms/epidemiology , Functional Laterality , Glioma/epidemiology , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Risk Factors , United Kingdom
6.
J Natl Compr Canc Netw ; 15(11): 1331-1345, 2017 11.
Article in English | MEDLINE | ID: mdl-29118226

ABSTRACT

For many years, the diagnosis and classification of gliomas have been based on histology. Although studies including large populations of patients demonstrated the prognostic value of histologic phenotype, variability in outcomes within histologic groups limited the utility of this system. Nonetheless, histology was the only proven and widely accessible tool available at the time, thus it was used for clinical trial entry criteria, and therefore determined the recommended treatment options. Research to identify molecular changes that underlie glioma progression has led to the discovery of molecular features that have greater diagnostic and prognostic value than histology. Analyses of these molecular markers across populations from randomized clinical trials have shown that some of these markers are also predictive of response to specific types of treatment, which has prompted significant changes to the recommended treatment options for grade III (anaplastic) gliomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Central Nervous System Neoplasms/diagnosis , Glioma/diagnosis , Nervous System/pathology , Antineoplastic Combined Chemotherapy Protocols/standards , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/therapy , Combined Modality Therapy/methods , Combined Modality Therapy/standards , Glioma/classification , Glioma/pathology , Glioma/therapy , Humans , Neoadjuvant Therapy/methods , Neoadjuvant Therapy/standards , Neoplasm Grading , Prognosis , Radiotherapy/methods , Radiotherapy/standards
7.
J Natl Compr Canc Netw ; 14(5 Suppl): 681-4, 2016 05.
Article in English | MEDLINE | ID: mdl-27226514

ABSTRACT

Over the past 30 years, the incidence of primary brain tumors has been increasing. Primary brain tumors are a heterogeneous group of central nervous system cancers with a wide range of outcomes and therapeutic strategies. As a result, prognostic features and molecular markers play a critical role in selecting treatment options for patients with brain tumors. At the NCCN 21st Annual Conference, Louis Burt Nabors, MD, reviewed the most appropriate adjuvant therapy for 3 types of brain tumors-primary low-grade gliomas, anaplastic oligodendrogliomas, and malignant gliomas-and shared supportive clinical trial data and NCCN recommendations.


Subject(s)
Central Nervous System Neoplasms/drug therapy , Glioma/drug therapy , Central Nervous System Neoplasms/pathology , Glioma/pathology , Humans
8.
Oncotarget ; 7(12): 15018-32, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26918452

ABSTRACT

Integrins αvß3 and αvß5 regulate angiogenesis and invasiveness in cancer, potentially by modulating activation of the transforming growth factor (TGF)-ß pathway. The randomized phase III CENTRIC and phase II CORE trials explored the integrin inhibitor cilengitide in patients with newly diagnosed glioblastoma with versus without O6-methylguanine DNA methyltransferase (MGMT) promoter methylation. These trials failed to meet their primary endpoints.Immunohistochemistry was used to assess the levels of the target integrins of cilengitide, αvß3 and αvß5 integrins, of αvß8 and of their putative target, phosphorylation of SMAD2, in tumor tissues from CENTRIC (n=274) and CORE (n=224).αvß3 and αvß5 expression correlated well in tumor and endothelial cells, but showed little association with αvß8 or pSMAD2 levels. In CENTRIC, there was no interaction between the biomarkers and treatment for prediction of outcome. In CORE, higher αvß3 levels in tumor cells were associated with improved progression-free survival by central review and with improved overall survival in patients treated with cilengitide.Integrins αvß3, αvß5 and αvß8 are differentially expressed in glioblastoma. Integrin levels do not correlate with the activation level of the canonical TGF-ß pathway. αvß3 integrin expression may predict benefit from integrin inhibition in patients with glioblastoma lacking MGMT promoter methylation.


Subject(s)
Biomarkers, Tumor/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Integrin alphaVbeta3/metabolism , Receptors, Vitronectin/metabolism , Smad2 Protein/metabolism , Snake Venoms/therapeutic use , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate , Young Adult
9.
J Natl Compr Canc Netw ; 13(10): 1191-202, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26483059

ABSTRACT

The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Central Nervous System (CNS) Cancers provide interdisciplinary recommendations for managing adult CNS cancers. Primary and metastatic brain tumors are a heterogeneous group of neoplasms with varied outcomes and management strategies. These NCCN Guidelines Insights summarize the NCCN CNS Cancers Panel's discussion and highlight notable changes in the 2015 update. This article outlines the data and provides insight into panel decisions regarding adjuvant radiation and chemotherapy treatment options for high-risk newly diagnosed low-grade gliomas and glioblastomas. Additionally, it describes the panel's assessment of new data and the ongoing debate regarding the use of alternating electric field therapy for high-grade gliomas.


Subject(s)
Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/radiotherapy , Practice Guidelines as Topic , Adult , Central Nervous System Neoplasms/pathology , Humans , Neoplasm Metastasis
10.
Biomolecules ; 5(1): 263-81, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25803745

ABSTRACT

The mRNA binding protein HuR is over expressed in cancer cells and contributes to disease progression through post-transcriptional regulation of mRNA. The regulation of HuR and how this relates to glioma is the focus of this report. SRC and c-Abl kinases regulate HuR sub-cellular trafficking and influence accumulation in the pericentriolar matrix (PCM) via a growth factor dependent signaling mechanism. Growth factor stimulation of glioma cell lines results in the associate of HuR with the PCM and amplification of centrosome number. This process is regulated by tyrosine phosphorylation of HuR and is abolished by mutating tyrosine residues. HuR is overexpressed in tumor samples from patients with glioblastoma and associated with a reduced survival. These findings suggest HuR plays a significant role in centrosome amplification and genomic instability, which contributes to a worse disease outcome.


Subject(s)
Centrosome/drug effects , Centrosome/metabolism , ELAV Proteins/metabolism , Genomic Instability/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Cell Line, Tumor , ELAV Proteins/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism , src-Family Kinases/metabolism
11.
J Natl Compr Canc Netw ; 12(11): 1517-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25361798

ABSTRACT

The NCCN Guidelines for Central Nervous System Cancers provide multidisciplinary recommendations for the clinical management of patients with cancers of the central nervous system. These NCCN Guidelines Insights highlight recent updates regarding the management of metastatic brain tumors using radiation therapy. Use of stereotactic radiosurgery (SRS) is no longer limited to patients with 3 or fewer lesions, because data suggest that total disease burden, rather than number of lesions, is predictive of survival benefits associated with the technique. SRS is increasingly becoming an integral part of management of patients with controlled, low-volume brain metastases.


Subject(s)
Central Nervous System Neoplasms/secondary , Central Nervous System Neoplasms/surgery , Humans , Radiosurgery/methods
12.
Lancet Oncol ; 15(10): 1100-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25163906

ABSTRACT

BACKGROUND: Cilengitide is a selective αvß3 and αvß5 integrin inhibitor. Data from phase 2 trials suggest that it has antitumour activity as a single agent in recurrent glioblastoma and in combination with standard temozolomide chemoradiotherapy in newly diagnosed glioblastoma (particularly in tumours with methylated MGMT promoter). We aimed to assess cilengitide combined with temozolomide chemoradiotherapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter. METHODS: In this multicentre, open-label, phase 3 study, we investigated the efficacy of cilengitide in patients from 146 study sites in 25 countries. Eligible patients (newly diagnosed, histologically proven supratentorial glioblastoma, methylated MGMT promoter, and age ≥18 years) were stratified for prognostic Radiation Therapy Oncology Group recursive partitioning analysis class and geographic region and centrally randomised in a 1:1 ratio with interactive voice response system to receive temozolomide chemoradiotherapy with cilengitide 2000 mg intravenously twice weekly (cilengitide group) or temozolomide chemoradiotherapy alone (control group). Patients and investigators were unmasked to treatment allocation. Maintenance temozolomide was given for up to six cycles, and cilengitide was given for up to 18 months or until disease progression or unacceptable toxic effects. The primary endpoint was overall survival. We analysed survival outcomes by intention to treat. This study is registered with ClinicalTrials.gov, number NCT00689221. FINDINGS: Overall, 3471 patients were screened. Of these patients, 3060 had tumour MGMT status tested; 926 patients had a methylated MGMT promoter, and 545 were randomly assigned to the cilengitide (n=272) or control groups (n=273) between Oct 31, 2008, and May 12, 2011. Median overall survival was 26·3 months (95% CI 23·8-28·8) in the cilengitide group and 26·3 months (23·9-34·7) in the control group (hazard ratio 1·02, 95% CI 0·81-1·29, p=0·86). None of the predefined clinical subgroups showed a benefit from cilengitide. We noted no overall additional toxic effects with cilengitide treatment. The most commonly reported adverse events of grade 3 or worse in the safety population were lymphopenia (31 [12%] in the cilengitide group vs 26 [10%] in the control group), thrombocytopenia (28 [11%] vs 46 [18%]), neutropenia (19 [7%] vs 24 [9%]), leucopenia (18 [7%] vs 20 [8%]), and convulsion (14 [5%] vs 15 [6%]). INTERPRETATION: The addition of cilengitide to temozolomide chemoradiotherapy did not improve outcomes; cilengitide will not be further developed as an anticancer drug. Nevertheless, integrins remain a potential treatment target for glioblastoma. FUNDING: Merck KGaA, Darmstadt, Germany.


Subject(s)
Brain Neoplasms/drug therapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Snake Venoms/therapeutic use , Tumor Suppressor Proteins/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Confidence Intervals , Dacarbazine/therapeutic use , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Early Detection of Cancer/methods , Female , Follow-Up Studies , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Patient Selection , Promoter Regions, Genetic , Proportional Hazards Models , Reference Values , Survival Analysis , Temozolomide , Treatment Outcome
13.
J Natl Compr Canc Netw ; 11(9): 1114-51, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24029126

ABSTRACT

Primary and metastatic tumors of the central nervous system are a heterogeneous group of neoplasms with varied outcomes and management strategies. Recently, improved survival observed in 2 randomized clinical trials established combined chemotherapy and radiation as the new standard for treating patients with pure or mixed anaplastic oligodendroglioma harboring the 1p/19q codeletion. For metastatic disease, increasing evidence supports the efficacy of stereotactic radiosurgery in treating patients with multiple metastatic lesions but low overall tumor volume. These guidelines provide recommendations on the diagnosis and management of this group of diseases based on clinical evidence and panel consensus. This version includes expert advice on the management of low-grade infiltrative astrocytomas, oligodendrogliomas, anaplastic gliomas, glioblastomas, medulloblastomas, supratentorial primitive neuroectodermal tumors, and brain metastases. The full online version, available at NCCN. org, contains recommendations on additional subtypes.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/therapy , Humans
14.
Clin Cancer Res ; 18(11): 3154-62, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22472174

ABSTRACT

PURPOSE: Glioblastoma is a devastating, incurable disease with few known prognostic factors. Here, we present the first genome-wide survival and validation study for glioblastoma. EXPERIMENTAL DESIGN: Cox regressions for survival with 314,635 inherited autosomal single-nucleotide polymorphisms (SNP) among 315 San Francisco Adult Glioma Study patients for discovery and three independent validation data sets [87 Mayo Clinic, 232 glioma patients recruited from several medical centers in Southeastern United States (GliomaSE), and 115 The Cancer Genome Atlas patients] were used to identify SNPs associated with overall survival for Caucasian glioblastoma patients treated with the current standard of care, resection, radiation, and temozolomide (total n = 749). Tumor expression of the gene that contained the identified prognostic SNP was examined in three separate data sets (total n = 619). Genotype imputation was used to estimate hazard ratios (HR) for SNPs that had not been directly genotyped. RESULTS: From the discovery and validation analyses, we identified a variant in single-stranded DNA-binding protein 2 (SSBP2) on 5q14.1 associated with overall survival in combined analyses (HR, 1.64; P = 1.3 × 10(-6)). Expression of SSBP2 in tumors from three independent data sets also was significantly related to patient survival (P = 5.3 × 10(-4)). Using genotype imputation, the SSBP2 SNP rs17296479 had the strongest statistically significant genome-wide association with poorer overall patient survival (HR, 1.79; 95% CI, 1.45-2.22; P = 1.0 × 10(-7)). CONCLUSION: The minor allele of SSBP2 SNP rs17296479 and the increased tumor expression of SSBP2 were statistically significantly associated with poorer overall survival among glioblastoma patients. With further confirmation, previously unrecognized inherited variations influencing survival may warrant inclusion in clinical trials to improve randomization. Unaccounted for genetic influence on survival could produce unwanted bias in such studies.


Subject(s)
Brain Neoplasms/genetics , DNA-Binding Proteins/genetics , Glioblastoma/genetics , Polymorphism, Single Nucleotide , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Genome-Wide Association Study , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Male , Middle Aged , Prognosis , Temozolomide
16.
Future Oncol ; 7(3): 339-54, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21417900

ABSTRACT

Cilengitide, a cyclicized arginine-glycine-aspartic acid-containing pentapeptide, potently blocks ανß3 and ανß5 integrin activation. Integrins are upregulated in many malignancies and mediate a wide variety of tumor-stroma interactions. Cilengitide and other integrin-targeting therapeutics have preclinical activity against many cancer subtypes including glioblastoma (GBM), the most common and deadliest CNS tumor. Cilengitide is active against orthotopic GBM xenografts and can augment radiotherapy and chemotherapy in these models. In Phase I and II GBM trials, cilengitide and the combination of cilengitide with standard temozolomide and radiation demonstrate consistent antitumor activity and a favorable safety profile. Cilengitide is currently under evaluation in a pivotal, randomized Phase III study (Cilengitide in Combination With Temozolomide and Radiotherapy in Newly Diagnosed Glioblastoma Phase III Randomized Clinical Trial [CENTRIC]) for newly diagnosed GBM. In addition, randomized controlled Phase II studies with cilengitide are ongoing for non-small-cell lung cancer and squamous cell carcinoma of the head and neck. Cilengitide is the first integrin inhibitor in clinical Phase III development for oncology.


Subject(s)
Antineoplastic Agents/therapeutic use , Glioblastoma/drug therapy , Integrins/antagonists & inhibitors , Oligopeptides/therapeutic use , Snake Venoms/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Industry/trends , Humans , Oligopeptides/adverse effects , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Snake Venoms/adverse effects , Snake Venoms/chemistry , Snake Venoms/pharmacokinetics , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...