Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
1.
Neurooncol Adv ; 4(1): vdac123, 2022.
Article in English | MEDLINE | ID: mdl-36225651

ABSTRACT

Background: Systemic therapies for refractory meningiomas are limited with no FDA-approved therapeutics. Vascular endothelial growth factor (VEGF) is a signaling protein associated with neovascularization, peritumoral edema, and meningioma tumorigenesis. Methods: This phase II study investigates the efficacy of bevacizumab (BEV), a VEGF binding monoclonal antibody, in patients with progressive Grade I (G1M), Grade II (G2M), Grade III (G3M) meningioma, and other non-parenchymal tumors including vestibular schwannoma (n = 4) and hemangiopericytoma (n = 4) with the primary endpoint of progression-free survival rate at 6-months (PFS-6). Non-meningiomas were included with the respective meningioma grade in the analysis. Secondary endpoints include median overall survival (mOS) and response rate. Results: Fifty Patients (26 women; median age 54 years; range 23-81), 42 with progressive meningioma were treated: 10 G1M, 20 G2M, and 12 G3M. Prior treatments include surgical resection (41 patients), radiosurgery (24 patients), external beam radiotherapy (28 patients), and chemotherapy (14 patients). Median infusions administered were 16 (range, 2-68). Response was graded using the Macdonald's criteria. PFS-6, median PFS, and mOS were 87%, 22 months, 35 months for G1M; 77%, 23 months, 41 months for G2M; and 46%, 8 months, 12 months for G3M. Best radiographic responses include stable disease (G1M: 100%; G2M: 85%; G3M: 82%); partial response (G1M: 0%; G2M: 5%; G3M: 0%) and progressive disease (G1M: 0%; G2M: 10%; G3M:18%). The most common toxicities were hypertension (n = 19, 42.2%), proteinuria (n = 16, 35.6%), and fatigue (n = 14, 31.1%). Conclusion: This study showed BEV is well tolerated and appears to be a promising systemic treatment option for patients with recurrent and refractory meningiomas.

2.
Neuro Oncol ; 24(1): 125-126, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34551114
4.
Neuro Oncol ; 20(7): 897-906, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29788429

ABSTRACT

Background: Corticosteroids are the mainstay of treatment for peritumor edema but are often associated with significant side effects. Therapies that can reduce corticosteroid use would potentially be of significant benefit to patients. However, currently there are no standardized endpoints evaluating corticosteroid use in neuro-oncology clinical trials. Methods: The Response Assessment in Neuro-Oncology (RANO) Working Group has developed consensus recommendations for endpoints evaluating corticosteroid use in clinical trials in both adults and children with brain tumors. Results: Responders are defined as patients with a 50% reduction in total daily corticosteroid dose compared with baseline or reduction of the total daily dose to ≤2 mg of dexamethasone (or equivalent dose of other corticosteroid); baseline dose must be at least 4 mg of dexamethasone daily (or equivalent dose of other corticosteroids) for at least one week. Patients must have stable or improved Neurologic Assessment in Neuro-Oncology (NANO) score or Karnofsky performance status score or Eastern Cooperative Oncology Group (ECOG) (Lansky score for children age <16 y), and an improved score on a relevant clinical outcome assessment tool. These criteria must be sustained for at least 4 weeks after baseline assessment to be considered a response, and are confirmed 4 weeks after that (ie, 8 wk after baseline assessment) to be considered a sustained response. Conclusions: This RANO proposal for corticosteroid use endpoints in neuro-oncology clinical trials may need to be refined and will require prospective validation in clinical studies.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Brain Edema/diagnostic imaging , Brain Neoplasms/complications , Neuroimaging/methods , Risk Assessment/methods , Brain Edema/drug therapy , Brain Edema/etiology , Brain Neoplasms/therapy , Humans
5.
Handb Clin Neurol ; 149: 169-204, 2018.
Article in English | MEDLINE | ID: mdl-29307353

ABSTRACT

Leptomeningeal metastasis (LM) results from dissemination of cancer cells to both the leptomeninges (pia and arachnoid) and cerebrospinal fluid (CSF) compartment. Breast cancer, lung cancer, and melanoma are the most common solid tumors that cause LM. Recent approval of more active anticancer therapies has resulted in improvement in survival that is partly responsible for an increased incidence of LM. Neurologic deficits, once manifest, are mostly irreversible, and often have a significant impact on patient quality of life. LM-directed therapy is based on symptom palliation, circumscribed use of neurosurgery, limited field radiotherapy, intra-CSF and systemic therapies. Novel methods of detecting LM include detection of CSF circulating tumor cells and tumor cell-free DNA. A recent international guideline for a standardization of response assessment in LM may improve cross-trial comparisons as well as within-trial evaluation of treatment. An increasing number of retrospective studies suggest that molecular-targeted therapy, such as EGFR and ALK inhibitors in lung cancer, trastuzumab in HER2+ breast cancer, and BRAF inhibitors in melanoma, may be effective as part of the multidisciplinary management of LM. Prospective randomized trials with standardized response assessment are needed to further validate these preliminary findings.


Subject(s)
Antineoplastic Agents/therapeutic use , Meningeal Neoplasms/drug therapy , Meningeal Neoplasms/secondary , Antineoplastic Agents/adverse effects , Breast Neoplasms/pathology , Female , Humans , Lung Neoplasms/pathology , Male , Melanoma/pathology , Meningeal Neoplasms/chemically induced
6.
Neuro Oncol ; 20(1): 13-23, 2018 01 10.
Article in English | MEDLINE | ID: mdl-28449033

ABSTRACT

Lack of standard response criteria in clinical trials for medulloblastoma and other seeding tumors complicates assessment of therapeutic efficacy and comparisons across studies. An international working group was established to develop consensus recommendations for response assessment. The aim is that these recommendations be prospectively evaluated in clinical trials, with the goal of achieving more reliable risk stratification and uniformity across clinical trials. Current practices and literature review were performed to identify major confounding issues and justify subsequently developed recommendations; in areas lacking scientific investigations, recommendations were based on experience of committee members and consensus was reached after discussion. Recommendations apply to both adult and pediatric patients with medulloblastoma and other seeding tumors. Response should be assessed using MR imaging (brain and spine), CSF cytology, and neurologic examination. Clinical imaging standards with minimum mandatory sequence acquisition that optimizes detection of leptomeningeal metastases are defined. We recommend central review prior to inclusion in treatment cohorts to ensure appropriate risk stratification and cohort inclusion. Consensus recommendations and response definitions for patients with medulloblastomas and other seeding tumors have been established; as with other Response Assessment in Neuro-Oncology recommendations, these need to now be prospectively validated in clinical trials.


Subject(s)
Brain Neoplasms , Medulloblastoma , Meningeal Neoplasms , Brain/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Child , Humans , Medulloblastoma/classification , Medulloblastoma/diagnostic imaging , Medulloblastoma/therapy , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/therapy , Neoplasm Seeding , Neuroimaging
7.
J Neurooncol ; 133(3): 561-569, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28500559

ABSTRACT

A single institution retrospective evaluation of nivolumab following disease progression on bevacizumab in adults with recurrent glioblastoma (GBM) with an objective of determining progression free survival (PFS). There is no accepted therapy for recurrent GBM after failure of bevacizumab. 16 adults, ages 52-72 years (median 62), with recurrent GBM were treated. All patients had previously been treated with surgery, concurrent radiotherapy and temozolomide, and post-radiotherapy temozolomide. Bevacizumab (with or without lomustine) was administered to all patients at first recurrence. Patients were treated with nivolumab only (3 mg/kg) once every 2 weeks at second recurrence. One cycle of nivolumab was defined as 2 treatments. Neurological evaluation was performed bi-weekly and neuroradiographic assessment every 4 weeks. A total of 37 treatment cycles (median 2) were administered of nivolumab in which there were 14 Grade 2 adverse events (AEs) and Grade 3 AEs in two patients. No Grade 4 or 5 AEs were seen. Following 1 month of nivolumab, seven patients demonstrated progressive disease and discontinued therapy. No patient demonstrated a response though nine patients demonstrated neuroradiographic stable response. Survival in the entire cohort ranged from 2 to 6 months with a median of 3.5 months (CI 2.8, 4.2). Median and 6-month PFS at 6 months was 2.0 months (range 1-5 months; CI 1.3, 2.7) and 0% respectively. Nivolumab salvage therapy demonstrated no survival advantage in patients with recurrent bevacizumab refractory GBM emphasizing a continued unmet need in neuro-oncology.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Bevacizumab/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Neoplasm Recurrence, Local/drug therapy , Aged , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Brain Neoplasms/diagnostic imaging , Disease Progression , Female , Glioblastoma/diagnostic imaging , Humans , Male , Middle Aged , Nivolumab , Retrospective Studies , Salvage Therapy/adverse effects , Survival Analysis , Treatment Outcome
8.
Cancer Control ; 24(1): S1-S16, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28557973

ABSTRACT

BACKGROUND: Neoplastic meningitis, also known as leptomeningeal disease, affects the entire neuraxis. The clinical manifestations of the disease may affect the cranial nerves, cerebral hemispheres, or the spine. Because of the extent of disease involvement, treatment options and disease staging should involve all compartments of the cerebrospinal fluid (CSF) and subarachnoid space. Few studies of patients with primary brain tumors have specifically addressed treatment for the secondary complication of neoplastic meningitis. Therapy for neoplastic meningitis is palliative in nature and, rarely, may have a curative intent. METHODS: A review of the medical literature pertinent to neoplastic meningitis in primary brain tumors was performed. The complication of neoplastic meningitis is described in detail for the various types of primary brain tumors. RESULTS: Treatment of neoplastic meningitis is complicated because determining who should receive aggressive, central nervous system (CNS)-directed therapy is difficult. In general, the therapeutic response of neoplastic meningitis is a function of CSF cytology and, secondarily, of the clinical improvement in neurological manifestations related to the disease. CSF cytology may manifest a rostrocaudal disassociation; thus, consecutive, negative findings require that both lumbar and ventricular cytological testing are performed to confirm the complete response. Based on data from several prospective, randomized trials extrapolated to primary brain tumors, the median rate of survival for neoplastic meningitis is several months. Oftentimes, therapy directed at palliation may improve quality of life by protecting patients from experiencing continued neurological deterioration. CONCLUSIONS: Neoplastic meningitis is a complicated disease in which response to therapy varies by histology. Thus, survival rates after CNS-directed therapy will differ by the underlying primary tumor. Optimal therapy of neoplastic meningitis is poorly defined, and few guidelines exist to guide clinicians on the most appropriate choice of therapy.


Subject(s)
Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/pathology , Cerebrospinal Fluid/cytology , Meningeal Neoplasms/cerebrospinal fluid , Meningeal Neoplasms/pathology , Meningitis/cerebrospinal fluid , Meningitis/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Combined Modality Therapy/methods , Humans , Meningeal Neoplasms/drug therapy , Meningeal Neoplasms/therapy , Meningitis/drug therapy , Meningitis/therapy , Prospective Studies , Quality of Life , Survival Rate
9.
J Neurooncol ; 131(3): 507-516, 2017 02.
Article in English | MEDLINE | ID: mdl-28204914

ABSTRACT

There is no standard therapy for recurrent anaplastic glioma (AG). Salvage therapies include alkylator-based chemotherapy, re-resection with or without carmustine implants, re-irradiation and bevacizumab. Bendamustine is a novel bifunctional alkylator with CNS penetration never previously evaluated in AG. Assess response and toxicity of bendamustine in recurrent AG in a phase II trial. Adults with radiation and temozolomide refractory recurrent AG were treated with bendamustine. A cycle of bendamustine was defined as two consecutive days of treatment (100 mg/m2/day) administered once every 4 weeks. Success of treatment was defined as progression free survival (PFS) at 6 months of 40 % or better. Twenty-six adults [16 males; 10 females: median age 40 years (range 30-65)] were treated, 12 at first recurrence and 17 at second recurrence. Prior salvage therapy included re-resection (14), chemotherapy (11) and re-radiation (2). Grade 3 treatment-related toxicities included lymphopenia (11 patients; Grade 4 in 3), myalgia, pneumonia, diarrhea, leukopenia, allergic reaction and thrombocytopenia in one patient each. One patient discontinued therapy due to toxicity. There were five instances of bendamustine dose delays all due to lymphopenia. There were no dose reductions due to toxicity. The median number of cycles of therapy was 3 (range 1-8). Best radiographic response was progressive disease in 12 (46 %), stable disease in 13 (50 %) and partial response in 1 (4 %). Median, 6- and 12-month PFS was 2.7 months (range 1-52), 27 and 8 % respectively. In patients with recurrent AG refractory to Z, bendamustine has manageable toxicity and modest single agent activity though not meeting pre-specified study criteria.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Bendamustine Hydrochloride/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Salvage Therapy , Adult , Aged , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prospective Studies , Temozolomide , Treatment Outcome
10.
Clin Lymphoma Myeloma Leuk ; 17(4): 225-230, 2017 04.
Article in English | MEDLINE | ID: mdl-28169156

ABSTRACT

INTRODUCTION: The purpose of our study was to identify the key risk factors for the development of posterior reversible encephalopathy syndrome (PRES) after administration of the combination chemotherapy regimen, DA-EPOCH (dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin). MATERIALS AND METHODS: We performed a retrospective medical record review of patients receiving DA-EPOCH with or without rituximab (DA-EPOCH ± R) at our institution from July 2012 to September 2014. The patients were screened for evidence of severe neurotoxicity through identification of requests for neurology consultations or neuroimaging studies. Patients with evidence of central nervous system (CNS) neurotoxicity were reviewed in detail to identify documented cases of PRES. The key risk factors assessed included rituximab administration sequence, and the presence of CNS insults, fluid status or electrolyte abnormalities, organ dysfunction, and hypertension. RESULTS: A total of 44 patients received DA-EPOCH ± R at our institution from July 2012 to September 2014. Of these 44 patients, 3 (7%) were diagnosed with PRES. The patients who developed PRES were more likely to have a pre-existing CNS insult, fluid status or electrolytes abnormalities, and hypertension. CONCLUSION: To the best of our knowledge, the present study is the first description of PRES associated with DA-EPOCH. The key risk factors for the development of PRES identified in our study included hypertension, fluid imbalance, electrolyte abnormalities, baseline organ dysfunction, a high tumor burden, and the presence of pre-existing CNS insults during chemotherapy, such as CNS infection. Patients with these risk factors appear to have a greater risk of developing PRES and should be monitored closely during treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Posterior Leukoencephalopathy Syndrome/chemically induced , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Central Nervous System/drug effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy , Prednisone/administration & dosage , Prednisone/adverse effects , Retrospective Studies , Risk Factors , Rituximab/administration & dosage , Vincristine/administration & dosage , Vincristine/adverse effects
11.
Cancer Control ; 24(1): 22-32, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28178709

ABSTRACT

BACKGROUND: Neoplastic meningitis, a central nervous system (CNS) complication of cancer metastatic to the meninges and cerebrospinal fluid (CSF), is relevant to oncologists due to the impact of the disease on patient quality of life and survival rates. METHODS: A review of the literature of articles published in English was conducted with regard to neoplastic meningitis. RESULTS: The incidence of neoplastic meningitis is increasing because patients with cancer are surviving longer in part because of the use of novel therapies with poor CNS penetration. Up to 5% of patients with solid tumors develop neoplastic meningitis during the disease course (breast cancer, lung cancer, and melanoma being the predominantly causative cancers). The rate of median survival in patients with untreated neoplastic meningitis is 1 to 2 months, although it can be as long as 5 months in some cases. Therapeutic options for the treatment of neoplastic meningitis include systemic therapy (cancer-specific, CNS-penetrating chemotherapy or targeted therapies), intra-CSF administration of chemotherapy (methotrexate, cytarabine, thiotepa) and CNS site-specific radiotherapy. Determining whom to treat with neoplastic meningitis remains challenging and, in part, relates to the extent of systemic disease, the neurological burden of disease, the available systemic therapies, and estimated rates of survival. CONCLUSIONS: The prognosis of neoplastic meningitis remains poor. The increasing use of novel, targeted therapies and immunotherapy in solid tumors and its impact on neoplastic meningitis remains to be determined and is an area of active research. Thus, well conducted trials are needed.


Subject(s)
Breast Neoplasms/complications , Lung Neoplasms/complications , Melanoma/complications , Meningitis/etiology , Breast Neoplasms/secondary , Combined Modality Therapy , Female , Humans , Lung Neoplasms/secondary , Melanoma/secondary , Meningitis/therapy , Prognosis , Severity of Illness Index , Survival Rate
12.
Neuro Oncol ; 19(1): i1-i24, 2017 01.
Article in English | MEDLINE | ID: mdl-28031389

ABSTRACT

Brain metastases (BM) occur frequently in many cancers, particularly non-small cell lung cancer (NSCLC), breast cancer, and melanoma. The development of BM is associated with poor prognosis and has an adverse impact on survival and quality of life. Commonly used therapies for BM such as surgery or radiotherapy are associated with only modest benefits. However, recent advances in systemic therapy of many cancers have generated considerable interest in exploration of those therapies for treatment of intracranial metastases.This review discusses the epidemiology of BM from the aforementioned primary tumors and the challenges of using systemic therapies for metastatic disease located within the central nervous system. Cumulative data from several retrospective and small prospective studies suggest that molecularly targeted systemic therapies may be an effective option for the treatment of BM from NSCLC, breast cancer, and melanoma, either as monotherapy or in conjunction with other therapies. Larger prospective studies are warranted to further characterize the efficacy and safety profiles of these targeted agents for the treatment of BM.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Melanoma/therapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Melanoma/pathology , Molecular Targeted Therapy
13.
CNS Oncol ; 5(4): 193-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27616612

ABSTRACT

ASCO 2016, 29 May-2 June 2016, Chicago, IL, USA The largest annual clinical oncology conference the American Society of Clinical Oncology is held in the USA and gives researchers and other key opinion leaders the opportunity to present new cancer clinical trials and research data. The CNS tumors section of the American Society of Clinical Oncology 2016 covered various aspects of neuro-oncology including metastatic CNS diseases and primary brain tumors, presented via posters, oral talks and over 100 abstracts. This brief review selectively highlights presentations from this meeting in an organizational manner that reflects clinically relevant aspects of a large and multifaceted meeting.


Subject(s)
Central Nervous System Neoplasms/therapy , Medical Oncology , Neurology , Central Nervous System Neoplasms/complications , Humans , Societies, Medical , United States
14.
Curr Neurol Neurosci Rep ; 16(9): 81, 2016 09.
Article in English | MEDLINE | ID: mdl-27443648

ABSTRACT

Chemotherapy may have detrimental effects on either the central or peripheral nervous system. Central nervous system neurotoxicity resulting from chemotherapy manifests as a wide range of clinical syndromes including acute, subacute, and chronic encephalopathies, posterior reversible encephalopathy, acute cerebellar dysfunction, chronic cognitive impairment, myelopathy, meningitis, and neurovascular syndromes. These clinical entities vary by causative agent, degree of severity, evolution, and timing of occurrence. In the peripheral nervous system, chemotherapy-induced peripheral neuropathy (CIPN) and myopathy are the two main complications of chemotherapy. CIPN is the most common complication, and the majority manifest as a dose-dependent length-dependent sensory axonopathy. In severe cases of CIPN, the dose of chemotherapy is reduced, the administration delayed, or the treatment discontinued. Few treatments are available for CIPN and based on meta-analysis, duloxetine is the preferred symptomatic treatment. Myopathy due to corticosteroid use is the most frequent cause of muscle disorders in patients with cancer.


Subject(s)
Peripheral Nervous System Diseases/chemically induced , Brain Diseases/chemically induced , Humans , Muscular Diseases/chemically induced , Neurotoxicity Syndromes/etiology
16.
CNS Oncol ; 5(3): 115-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27230972

ABSTRACT

20th Annual Meeting of the Society for Neuro-Oncology, San Antonio, TX, USA, 18-22 November 2015 The Society for Neuro-Oncology is the largest neuro-oncology meeting in the USA that meets annually and provides a multiday venue that showcases new brain cancer clinical trial results and basic research primarily pertaining to gliomas. The Society for Neuro-Oncology 2015 meeting comprising one education day, 2 days of premeetings and 3 days of presentation, over 200 oral presentations and 900 abstracts provides an overview of contemporary neuro-oncology that includes metastatic disease of the central nervous system as well as primary brain tumors. This review attempts to highlight select abstracts presented at this year's meeting in a short summary that provides a synopsis of a large and multifaceted meeting.

17.
CNS Oncol ; 5(3): 145-57, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27230974

ABSTRACT

Anaplastic astrocytoma (AA) is a diffusely infiltrating, malignant, astrocytic, primary brain tumor. AA is currently defined by histology although future classification schemes will include molecular alterations. AA can be separated into subgroups, which share similar molecular profiles, age at diagnosis and median survival, based on 1p/19q co-deletion status and IDH mutation status. AA with co-deletion of chromosomes 1p and 19q and IDH mutation have the best prognosis. AA with IDH mutation and no 1p/19q co-deletion have intermediate prognosis and AA with wild-type IDH have the worst prognosis and share many molecular alterations with glioblastoma. Treatment of noncodeleted AA based on preliminary results from the CATNON clinical trial consists of maximal safe resection followed by radiotherapy with post-radiotherapy temozolomide (TMZ) chemotherapy. The role of concurrent TMZ and whether IDH1 subgroups benefit from TMZ is currently being evaluated in the recently completed randomized, prospective Phase III clinical trial, CATNON.


Subject(s)
Astrocytoma , Brain Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Astrocytoma/diagnostic imaging , Astrocytoma/genetics , Astrocytoma/therapy , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Humans , Isocitrate Dehydrogenase/genetics , Mutation/genetics
19.
Neuro Oncol ; 18(9): 1199-208, 2016 09.
Article in English | MEDLINE | ID: mdl-27106405

ABSTRACT

This guideline provides recommendations for the use of PET imaging in gliomas. The review examines established clinical benefit in glioma patients of PET using glucose ((18)F-FDG) and amino acid tracers ((11)C-MET, (18)F-FET, and (18)F-FDOPA). An increasing number of studies have been published on PET imaging in the setting of diagnosis, biopsy, and resection as well radiotherapy planning, treatment monitoring, and response assessment. Recommendations are based on evidence generated from studies which validated PET findings by histology or clinical course. This guideline emphasizes the clinical value of PET imaging with superiority of amino acid PET over glucose PET and provides a framework for the use of PET to assist in the management of patients with gliomas.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioma/diagnostic imaging , Molecular Imaging/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Glioma/metabolism , Glioma/therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...