15451
|
Jiang H, McCormick F, Lang FF, Gomez-Manzano C, Fueyo J. Oncolytic adenoviruses as antiglioma agents. Expert Rev Anticancer Ther 2006; 6:697-708. [PMID: 16759161 DOI: 10.1586/14737140.6.5.697] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The treatment for malignant gliomas is suboptimal. Oncolytic adenoviruses hold the promise of being effective agents for the treatment of solid tumors. Importantly, the first oncolytic viral therapy has just been approved for use in combination with chemotherapy for late-stage refractory nasopharyngeal cancer by the Chinese State FDA, following a successful Phase III randomized clinical trial. The concept underlying treatment with oncolytic adenoviruses is based on cancer selectivity by confining viral replication and infectivity to cancer cells. For this purpose, the main strategies used currently to modify the viruses include: functional deletions in essential viral genes; tumor- or tissue-specific promoters used to control the expression of these viral genes; and tropism modification to redirect adenovirus to the cancer cell surface. In the near future, oncolytic adenoviruses need to be optimized to fully realize their potential as critical anticancer tools and, thus, improve the prognosis for patients with malignant gliomas.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Box 316, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
15452
|
Maier-Hauff K, Rothe R, Scholz R, Gneveckow U, Wust P, Thiesen B, Feussner A, von Deimling A, Waldoefner N, Felix R, Jordan A. Intracranial thermotherapy using magnetic nanoparticles combined with external beam radiotherapy: results of a feasibility study on patients with glioblastoma multiforme. J Neurooncol 2006; 81:53-60. [PMID: 16773216 DOI: 10.1007/s11060-006-9195-0] [Citation(s) in RCA: 406] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2006] [Accepted: 05/09/2006] [Indexed: 11/26/2022]
Abstract
We aimed to evaluate the feasibility and tolerability of the newly developed thermotherapy using magnetic nanoparticles on recurrent glioblastoma multiforme. Fourteen patients received 3-dimensional image guided intratumoral injection of aminosilane coated iron oxide nanoparticles. The patients were then exposed to an alternating magnetic field to induce particle heating. The amount of fluid and the spatial distribution of the depots were planned in advance by means of a specially developed treatment planning software following magnetic resonance imaging (MRI). The actually achieved magnetic fluid distribution was measured by computed tomography (CT), which after matching to pre-operative MRI data enables the calculation of the expected heat distribution within the tumor in dependence of the magnetic field strength. Patients received 4-10 (median: 6) thermotherapy treatments following instillation of 0.1-0.7 ml (median: 0.2) of magnetic fluid per ml tumor volume and single fractions (2 Gy) of a radiotherapy series of 16-70 Gy (median: 30). Thermotherapy using magnetic nanoparticles was tolerated well by all patients with minor or no side effects. Median maximum intratumoral temperatures of 44.6 degrees C (42.4-49.5 degrees C) were measured and signs of local tumor control were observed. In conclusion, deep cranial thermotherapy using magnetic nanoparticles can be safely applied on glioblastoma multiforme patients.
Collapse
|
15453
|
Hunter C, Smith R, Cahill DP, Stephens P, Stevens C, Teague J, Greenman C, Edkins S, Bignell G, Davies H, O’Meara S, Parker A, Avis T, Barthorpe S, Brackenbury L, Buck G, Butler A, Clements J, Cole J, Dicks E, Forbes S, Gorton M, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jenkinson A, Jones D, Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R, Raine K, Richardson D, Shepherd R, Small A, Solomon H, Tofts C, Varian J, West S, Widaa S, Yates A, Easton DF, Riggins G, Roy JE, Levine KK, Mueller W, Batchelor TT, Louis DN, Stratton MR, Andrew Futreal P, Wooster R. A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy. Cancer Res 2006; 66:3987-91. [PMID: 16618716 PMCID: PMC7212022 DOI: 10.1158/0008-5472.can-06-0127] [Citation(s) in RCA: 347] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Malignant gliomas have a very poor prognosis. The current standard of care for these cancers consists of extended adjuvant treatment with the alkylating agent temozolomide after surgical resection and radiotherapy. Although a statistically significant increase in survival has been reported with this regimen, nearly all gliomas recur and become insensitive to further treatment with this class of agents. We sequenced 500 kb of genomic DNA corresponding to the kinase domains of 518 protein kinases in each of nine gliomas. Large numbers of somatic mutations were observed in two gliomas recurrent after alkylating agent treatment. The pattern of mutations in these cases showed strong similarity to that induced by alkylating agents in experimental systems. Further investigation revealed inactivating somatic mutations of the mismatch repair gene MSH6 in each case. We propose that inactivating somatic mutations of MSH6 confer resistance to alkylating agents in gliomas in vivo and concurrently unleash accelerated mutagenesis in resistant clones as a consequence of continued exposure to alkylating agents in the presence of defective mismatch repair. The evidence therefore suggests that when MSH6 is inactivated in gliomas, alkylating agents convert from induction of tumor cell death to promotion of neoplastic progression. These observations highlight the potential of large scale sequencing for revealing and elucidating mutagenic processes operative in individual human cancers.
Collapse
Affiliation(s)
- Chris Hunter
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Raffaella Smith
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Daniel P. Cahill
- Molecular Pathology Unit, Brain Tumor Center, Neurosurgical Service and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Philip Stephens
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Claire Stevens
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jon Teague
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Chris Greenman
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Sarah Edkins
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Graham Bignell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Helen Davies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Sarah O’Meara
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Adrian Parker
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Tim Avis
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Syd Barthorpe
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Lisa Brackenbury
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Gemma Buck
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Adam Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jody Clements
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jennifer Cole
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Ed Dicks
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Simon Forbes
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Matthew Gorton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Kristian Gray
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Kelly Halliday
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Rachel Harrison
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Katy Hills
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jonathon Hinton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Andy Jenkinson
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - David Jones
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Vivienne Kosmidou
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Ross Laman
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Richard Lugg
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Andrew Menzies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Janet Perry
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Robert Petty
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Keiran Raine
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - David Richardson
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Rebecca Shepherd
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Alexandra Small
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Helen Solomon
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Calli Tofts
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jennifer Varian
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Sofie West
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Sara Widaa
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Andy Yates
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Douglas F. Easton
- Cancer Research UK Genetic Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Gregory Riggins
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer E. Roy
- Molecular Pathology Unit, Brain Tumor Center, Neurosurgical Service and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kymberly K. Levine
- Molecular Pathology Unit, Brain Tumor Center, Neurosurgical Service and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wolf Mueller
- Molecular Pathology Unit, Brain Tumor Center, Neurosurgical Service and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tracy T. Batchelor
- Molecular Pathology Unit, Brain Tumor Center, Neurosurgical Service and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - David N. Louis
- Molecular Pathology Unit, Brain Tumor Center, Neurosurgical Service and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michael R. Stratton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
- Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - P. Andrew Futreal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Richard Wooster
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
15454
|
Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F, Reulen HJ. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol 2006; 7:392-401. [PMID: 16648043 DOI: 10.1016/s1470-2045(06)70665-9] [Citation(s) in RCA: 2378] [Impact Index Per Article: 125.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND 5-Aminolevulinic acid is a non-fluorescent prodrug that leads to intracellular accumulation of fluorescent porphyrins in malignant gliomas-a finding that is under investigation for intraoperative identification and resection of these tumours. We aimed to assess the effect of fluorescence-guided resection with 5-aminolevulinic acid on surgical radicality, progression-free survival, overall survival, and morbidity. METHODS 322 patients aged 23-73 years with suspected malignant glioma amenable to complete resection of contrast-enhancing tumour were randomly assigned to 20 mg/kg bodyweight 5-aminolevulinic acid for fluorescence-guided resection (n=161) or to conventional microsurgery with white light (n=161). The primary endpoints were the number of patients without contrast-enhancing tumour on early MRI (ie, that obtained within 72 h after surgery) and 6-month progression-free survival as assessed by MRI. Secondary endpoints were volume of residual tumour on postoperative MRI, overall survival, neurological deficit, and toxic effects. We report the results of an interim analysis with 270 patients in the full-analysis population (139 assigned 5-aminolevulinic acid, 131 assigned white light), which excluded patients with ineligible histological and radiological findings as assessed by central reviewers who were masked as to treatment allocation; the interim analysis resulted in termination of the study as defined by the protocol. Primary and secondary endpoints were analysed by intention to treat in the full-analysis population. The study is registered at http://www.clinicaltrials.gov as NCT00241670. FINDINGS Median follow-up was 35.4 months (95% CI 1.0-56.7). Contrast-enhancing tumour was resected completely in 90 (65%) of 139 patients assigned 5-aminolevulinic acid compared with 47 (36%) of 131 assigned white light (difference between groups 29% [95% CI 17-40], p<0.0001). Patients allocated 5-aminolevulinic acid had higher 6-month progression free survival than did those allocated white light (41.0% [32.8-49.2] vs 21.1% [14.0-28.2]; difference between groups 19.9% [9.1-30.7], p=0.0003, Z test). Groups did not differ in the frequency of severe adverse events or adverse events in any organ system class reported within 7 days after surgery. INTERPRETATION Tumour fluorescence derived from 5-aminolevulinic acid enables more complete resections of contrast-enhancing tumour, leading to improved progression-free survival in patients with malignant glioma.
Collapse
Affiliation(s)
- Walter Stummer
- Neurochirurgische Klinik, Heinrich-Heine University, Dusseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
15455
|
Marosi C. Chemotherapy for malignant gliomas. Wien Med Wochenschr 2006; 156:346-50. [PMID: 16944366 DOI: 10.1007/s10354-006-0307-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Accepted: 03/31/2006] [Indexed: 11/29/2022]
Abstract
Concomitant and adjuvant treatment with Temozolomide, an oral alkylating agent, has significantly improved the survival of patients with newly diagnosed glioblastoma multiforme (study EORTC 26981/22981, NCIC CE3). When given with the appropriate cautiousness including weekly clinical and laboratory controls during the concomitant phase, this therapy is generally well tolerated. The observed toxicity is mainly haematological. Grade III and IV toxicities mainly thrombocytopenia or lymphocytopenia occur in around 5 % of patients. A prophylaxis against pneumocystis carinii pneumonia was mandatory in the EORTC study. Most importantly, the quality of life of the patients was maintained throughout the therapy. This success has boosted the whole field of neurooncology, after a dry spell of more than thirty years for glioblastoma multiforme. Whether this concept will be applicable to other brain tumours and which schedule modifications or combinations with biologicals will improve the effectivity of therapy in brain tumours should be explored in further studies.
Collapse
Affiliation(s)
- Christine Marosi
- Division of Medical Oncology, Department of Internal Medicine I, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
15456
|
Abstract
The dogma that the genesis of new cells is a negligible event in the adult mammalian brain has long influenced our perception and understanding of the origin and development of CNS tumours. The discovery that new neurons and glia are produced throughout life from neural stem cells provides new possibilities for the candidate cells of origin of CNS neoplasias. The emerging hypothesis is that alterations in the cellular and genetic mechanisms that control adult neurogenesis might contribute to brain tumorigenesis, thereby allowing the identification of new therapeutic strategies.
Collapse
Affiliation(s)
- Angelo L Vescovi
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan 20126, Italy.
| | | | | |
Collapse
|
15457
|
Mirimanoff RO, Gorlia T, Mason W, Van den Bent MJ, Kortmann RD, Fisher B, Reni M, Brandes AA, Curschmann J, Villa S, Cairncross G, Allgeier A, Lacombe D, Stupp R. Radiotherapy and Temozolomide for Newly Diagnosed Glioblastoma: Recursive Partitioning Analysis of the EORTC 26981/22981-NCIC CE3 Phase III Randomized Trial. J Clin Oncol 2006; 24:2563-9. [PMID: 16735709 DOI: 10.1200/jco.2005.04.5963] [Citation(s) in RCA: 364] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The European Organisation for Research and Treatment of Cancer and National Cancer Institute of Canada trial on temozolomide (TMZ) and radiotherapy (RT) in glioblastoma (GBM) has demonstrated that the combination of TMZ and RT conferred a significant and meaningful survival advantage compared with RT alone. We evaluated in this trial whether the recursive partitioning analysis (RPA) retains its overall prognostic value and what the benefit of the combined modality is in each RPA class. Patients and Methods Five hundred seventy-three patients with newly diagnosed GBM were randomly assigned to standard postoperative RT or to the same RT with concomitant TMZ followed by adjuvant TMZ. The primary end point was overall survival. The European Organisation for Research and Treatment of Cancer RPA used accounts for age, WHO performance status, extent of surgery, and the Mini-Mental Status Examination. Results Overall survival was statistically different among RPA classes III, IV, and V, with median survival times of 17, 15, and 10 months, respectively, and 2-year survival rates of 32%, 19%, and 11%, respectively (P < .0001). Survival with combined TMZ/RT was higher in RPA class III, with 21 months median survival time and a 43% 2-year survival rate, versus 15 months and 20% for RT alone (P = .006). In RPA class IV, the survival advantage remained significant, with median survival times of 16 v 13 months, respectively, and 2-year survival rates of 28% v 11%, respectively (P = .0001). In RPA class V, however, the survival advantage of RT/TMZ was of borderline significance (P = .054). Conclusion RPA retains its prognostic significance overall as well as in patients receiving RT with or without TMZ for newly diagnosed GBM, particularly in classes III and IV.
Collapse
|
15458
|
Rong Y, Durden DL, Van Meir EG, Brat DJ. ‘Pseudopalisading’ Necrosis in Glioblastoma: A Familiar Morphologic Feature That Links Vascular Pathology, Hypoxia, and Angiogenesis. J Neuropathol Exp Neurol 2006; 65:529-39. [PMID: 16783163 DOI: 10.1097/00005072-200606000-00001] [Citation(s) in RCA: 380] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma (GBM) is a highly malignant, rapidly progressive astrocytoma that is distinguished pathologically from lower grade tumors by necrosis and microvascular hyperplasia. Necrotic foci are typically surrounded by "pseudopalisading" cells-a configuration that is relatively unique to malignant gliomas and has long been recognized as an ominous prognostic feature. Precise mechanisms that relate morphology to biologic behavior have not been described. Recent investigations have demonstrated that pseudopalisades are severely hypoxic, overexpress hypoxia-inducible factor (HIF-1), and secrete proangiogenic factors such as VEGF and IL-8. Thus, the microvascular hyperplasia in GBM that provides a new vasculature and promotes peripheral tumor expansion is tightly linked with the emergence of pseudopalisades. Both pathologic observations and experimental evidence have indicated that the development of hypoxia and necrosis within astrocytomas could arise secondary to vaso-occlusion and intravascular thrombosis. This emerging model suggests that pseudopalisades represent a wave of tumor cells actively migrating away from central hypoxia that arises after a vascular insult. Experimental glioma models have shown that endothelial apoptosis, perhaps resulting from angiopoetin-2, initiates vascular pathology, whereas observations in human tumors have clearly demonstrated that intravascular thrombosis develops with high frequency in the transition to GBM. Tissue factor, the main cellular initiator of thrombosis, is dramatically upregulated in response to PTEN loss and hypoxia in human GBM and could promote a prothrombotic environment that precipitates these events. A prothrombotic environment also activates the family of protease activated receptors (PARs) on tumor cells, which are G-protein-coupled and enhance invasive and proangiogenic properties. Vaso-occlusive and prothrombotic mechanisms in GBM could readily explain the presence of pseudopalisading necrosis in tissue sections, the rapid peripheral expansion on neuroimaging, and the dramatic shift to an accelerated rate of clinical progression resulting from hypoxia-induced angiogenesis.
Collapse
Affiliation(s)
- Yuan Rong
- Department of Pathology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
15459
|
Preusser M, Haberler C, Hainfellner JA. Malignant glioma: Neuropathology and Neurobiology. Wien Med Wochenschr 2006; 156:332-7. [PMID: 16944363 DOI: 10.1007/s10354-006-0304-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Accepted: 03/31/2006] [Indexed: 12/17/2022]
Abstract
Malignant gliomas may manifest at any age including congenital and childhood cases. Peak incidence is, however, in adults older than 40 years. Males are more frequently affected than females. The sole unequivocal risk factor is therapeutic ionizing irradiation. Malignant gliomas comprise a spectrum of different tumor subtypes. Within this spectrum, glioblastoma, anaplastic astrocytoma and anaplastic oligodendroglioma share as basic features preferential location in cerebral hemispheres, diffuse infiltration of brain tissue, fast tumor growth with fatal outcome within months or years. Invasion is regarded as one of the main reasons for poor therapeutic success, because it makes complete surgical removal of gliomas impossible. Invasion of glioma cells requires interaction with the extracellular matrix and with surrounding cells of the healthy brain tissue. Vascular proliferates and tissue necrosis are characteristic features of malignant gliomas, in particular glioblastoma. These features are most likely the consequence of rapidly increasing tumor mass that is inadequately oxygenized by the preexisting vasculature. In malignant glioma, distinct molecular pathways including the p53 pathway, the RB pathway and the EGFR pathway show frequent alterations that seem to be pathogenetically relevant. Methylguanine-methyltransferase (MGMT) promoter methylation status in glioblastoma and 1p19q deletion status in anaplastic oligodendroglioma are associated with response to chemotherapy. The role of neuropathology and neurobiology in neurooncology is 1. to provide a clinically meaningful classification of brain tumors on basis of pathobiological factors, 2. to clarify etiology and pathogenesis of brain tumors as rational basis for development of new diagnostic tests and therapies, and 3. to translate testing for new clinically relevant molecular parameters into clinical application.
Collapse
Affiliation(s)
- Matthias Preusser
- Institute of Neurology, Medical University Vienna, Währinger Gürtel 18-20, 1097 Vienna, Austria
| | | | | |
Collapse
|
15460
|
Bracard S, Taillandier L, Antoine V, Kremer S, Taillandier C, Schmitt E. [Cerebral gliomas: imaging diagnosis and follow-up]. JOURNAL DE RADIOLOGIE 2006; 87:779-91. [PMID: 16778747 DOI: 10.1016/s0221-0363(06)74087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The management of gliomas evolves towards more aggressive strategies with a combination of surgery, radiotherapy and chemotherapy. Follow-up imaging based on morphological MRI, with simple and reproducible protocols, may be associated with functional MRI and spectroscopy. Baseline postsurgical MRI must be performed within the first three days. Follow-up examinations should be done 2 months after radiotherapy and during chemotherapy, usually after each cycle of two or three treatments. Continued follow-up after therapy is recommended to assess response and detect recurrences or therapeutic complications.
Collapse
Affiliation(s)
- S Bracard
- Service de Neuroradiologie, Hôpital Central, CHU de Nancy, 29, avenue Maréchal-de-Lattre-de-Tassigny, 54035 Nancy.
| | | | | | | | | | | |
Collapse
|
15461
|
Hamilton DA. Adding concomitant and adjuvant temozolomide to radiotherapy does not reduce health-related quality of life in people with glioblastoma. Cancer Treat Rev 2006; 32:483-6. [PMID: 16730911 DOI: 10.1016/j.ctrv.2006.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- David A Hamilton
- Blood and Cancer Centre, Wellington Hospital, Wellington, New Zealand
| |
Collapse
|
15462
|
Wibom C, Pettersson F, Sjöström M, Henriksson R, Johansson M, Bergenheim AT. Protein expression in experimental malignant glioma varies over time and is altered by radiotherapy treatment. Br J Cancer 2006; 94:1853-63. [PMID: 16736004 PMCID: PMC2361353 DOI: 10.1038/sj.bjc.6603190] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Radiotherapy is one of the mainstays of glioblastoma (GBM) treatment. This study aims to investigate and characterise differences in protein expression patterns in brain tumour tissue following radiotherapy, in order to gain a more detailed understanding of the biological effects. Rat BT4C glioma cells were implanted into the brain of two groups of 12 BDIX-rats. One group received radiotherapy (12 Gy single fraction). Protein expression in normal and tumour brain tissue, collected at four different time points after irradiation, were analysed using surface enhanced laser desorption/ionisation – time of flight – mass spectrometry (SELDI-TOF-MS). Mass spectrometric data were analysed by principal component analysis (PCA) and partial least squares (PLS). Using these multivariate projection methods we detected differences between tumours and normal tissue, radiation treatment-induced changes and temporal effects. 77 peaks whose intensity significantly changed after radiotherapy were discovered. The prompt changes in the protein expression following irradiation might help elucidate biological events induced by radiation. The combination of SELDI-TOF-MS with PCA and PLS seems to be well suited for studying these changes. In a further perspective these findings may prove to be useful in the development of new GBM treatment approaches.
Collapse
Affiliation(s)
- C Wibom
- Department of Oncology, University Hospital, SE 901 85 Umeå, Sweden
| | - F Pettersson
- Research Group for Chemometrics, Department of Chemistry, Umeå University, SE 901 87 Umeå, Sweden
| | - M Sjöström
- Research Group for Chemometrics, Department of Chemistry, Umeå University, SE 901 87 Umeå, Sweden
| | - R Henriksson
- Department of Oncology, University Hospital, SE 901 85 Umeå, Sweden
| | - M Johansson
- Department of Oncology, University Hospital, SE 901 85 Umeå, Sweden
| | - A T Bergenheim
- Department of Neurosurgery, University Hospital, SE 901 85, Umeå, Sweden
- E-mail:
| |
Collapse
|
15463
|
Eyüpoglu IY, Hahnen E, Tränkle C, Savaskan NE, Siebzehnrübl FA, Buslei R, Lemke D, Wick W, Fahlbusch R, Blümcke I. Experimental therapy of malignant gliomas using the inhibitor of histone deacetylase MS-275. Mol Cancer Ther 2006; 5:1248-55. [PMID: 16731757 DOI: 10.1158/1535-7163.mct-05-0533] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inhibitors of histone deacetylases are promising compounds for the treatment of cancer but have not been systematically explored in malignant brain tumors. Here, we characterize the benzamide MS-275, a class I histone deacetylase inhibitor, as potent drug for experimental therapy of glioblastomas. Treatment of four glioma cell lines (U87MG, C6, F98, and SMA-560) with MS-275 significantly reduced cell growth in a concentration-dependent manner (IC(90), 3.75 micromol/L). Its antiproliferative effect was corroborated using a bromodeoxyuridine proliferation assay and was mediated by G(0)-G(1) cell cycle arrest (i.e., up-regulation of p21/WAF) and apoptotic cell death. Implantation of enhanced green fluorescent protein-transfected F98 glioma cells into slice cultures of rat brain confirmed the cytostatic effect of MS-275 without neurotoxic damage to the organotypic neuronal environment in a dose escalation up to 20 micromol/L. A single intratumoral injection of MS-275 7 days after orthotopic implantation of glioma cells in syngeneic rats confirmed the chemotherapeutic efficacy of MS-275 in vivo. Furthermore, its propensity to pass the blood-brain barrier and to increase the protein level of acetylated histone H3 in brain tissue identifies MS-275 as a promising candidate drug in the treatment of malignant gliomas.
Collapse
Affiliation(s)
- Ilker Y Eyüpoglu
- Department of Neurosurgery, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15464
|
Noronha V, Berliner N, Ballen KK, Lacy J, Kracher J, Baehring J, Henson JW. Treatment-related myelodysplasia/AML in a patient with a history of breast cancer and an oligodendroglioma treated with temozolomide: case study and review of the literature. Neuro Oncol 2006; 8:280-3. [PMID: 16728498 PMCID: PMC1871950 DOI: 10.1215/15228517-2006-003] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The emergence of temozolomide as an effective alkylating agent with little acute toxicity or cumulative myelosuppression has led to protracted courses of chemotherapy for many patients with gliomas. Secondary, or treatment-related, myelodysplasia (t-MDS) and acute myelogenous leukemia (t-AML) are life-threatening complications of alkylating chemotherapy and have been reported in patients with primary brain tumors. We describe a case of temozolomide-related t-MDS/AML and discuss the clinical features of this condition. Administration of an alkylating agent in patient populations with long median survivals must be undertaken with an understanding of the potential for this treatment complication.
Collapse
Affiliation(s)
| | | | | | | | | | | | - John W. Henson
- Address correspondence to John W. Henson, Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Yawkey 9 East, Fruit Street, Boston, MA 02114 (
)
| |
Collapse
|
15465
|
Brandes AA, Nicolardi L, Tosoni A, Gardiman M, Iuzzolino P, Ghimenton C, Reni M, Rotilio A, Sotti G, Ermani M. Survival following adjuvant PCV or temozolomide for anaplastic astrocytoma. Neuro Oncol 2006; 8:253-60. [PMID: 16723632 PMCID: PMC1871946 DOI: 10.1215/15228517-2006-005] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We compared survival in patients with anaplastic astrocytoma (AA) treated with adjuvant procarbazine, lomustine, and vincristine (PCV) with survival in patients treated with temozolomide. A retrospective analysis was made of patients with newly diagnosed AA treated with adjuvant postradiotherapy chemotherapy. Outcome analysis included progression-free survival and overall survival. The following prognostic factors were taken into account: patient age, extent of resection, performance status, presence of contrast enhancement in presurgical imaging, and type of adjuvant treatment. Among 109 AA patients, 49 were treated with PCV and 60 with temozolomide. The treatment groups were well matched for pretreatment characteristics, except for the presence of contrast enhancement. Age, extent of surgery, performance status, and presence of contrast enhancement were statistically significant prognostic factors according to the Cox model analysis of survival. Type of adjuvant chemotherapy was not a significant factor, either for progression-free survival or for overall survival. Hematological toxicity, nonhematological toxicity grades 3-4, and premature discontinuation due to toxicity were observed in 9%, 3% to 5%, and 37%, respectively, of cases in the PCV group versus 4% to 5%, 0, and 0, respectively, in the temozolomide group. Although the present study was not randomized, it was well designed, and it reports on two homogeneous and consecutive series of patients, for whom histology was verified to obtain survival data only for patients with AA following the recent WHO 2000 classification. Even if no survival advantage has been demonstrated for temozolomide versus PCV, we conclude that temozolomide should be preferred because of its greater tolerability.
Collapse
Affiliation(s)
- Alba A Brandes
- Department of Medical Oncology, Istituto Oncologico Veneto-IRCCS Padova, Padova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15466
|
Tang P, Roldan G, Brasher PMA, Fulton D, Roa W, Murtha A, Cairncross JG, Forsyth PA. A phase II study of carboplatin and chronic high-dose tamoxifen in patients with recurrent malignant glioma. J Neurooncol 2006; 78:311-6. [PMID: 16710748 DOI: 10.1007/s11060-005-9104-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Accepted: 12/07/2005] [Indexed: 11/25/2022]
Abstract
PURPOSE To determine the response rate, time to disease progression, survival, and toxicity of intravenous carboplatin and chronic oral high-dose tamoxifen in patients with recurrent malignant gliomas. PATIENTS AND METHODS Patients with histological confirmation of recurrent malignant gliomas were eligible for this multicenter phase II trial. Treatment consisted of 400 mg/m2 carboplatin intravenously every 4 weeks and oral high dose chronic tamoxifen (80 mg bid in women and 100 mg bid in men). RESULTS Twenty seven patients met the eligibility criteria and were evaluable for response. The histological subtypes were: 16 (59%) glioblastoma multiforme (GBM), malignant astrocytoma (5 patients), malignant mixed glioma (5 patients), and glioblastoma/gliosarcoma (1 patient). Twenty-two patients (82%) had an ECOG performance status of 0 or 1. No complete responses were observed, 4 patients (15%) achieved a partial response, and 14 patients (52%) had stable disease. Median time to progression was 3.65 months (95%CI 2.56, 4.83). Median overall survival was 14.09 months (95%CI 7.06, 19.91). One patient with a recurrent GBM had a sustained partial response and is progression free 81 months since starting treatment. Another patient with mixed malignant oligoastrocytoma also had a prolonged partial response (lasting 63 months) and is alive 84 months after treatment for recurrence. The most frequently reported grade 3 or 4 toxicities were fatigue (19%), nausea (11%) and anorexia (11%). CONCLUSIONS Carboplatin and high dose tamoxifen has similar response rates to other regimens for recurrent malignant gliomas and are probably equivalent to those found using tamoxifen as monotherapy. Long-lasting periods of disease free survival in some patients (particularly those with malignant mixed oligo astrocytomas) were found.
Collapse
Affiliation(s)
- P Tang
- Department of Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
15467
|
Combs SE, Heeger S, Haselmann R, Edler L, Debus J, Schulz-Ertner D. Treatment of primary glioblastoma multiforme with cetuximab, radiotherapy and temozolomide (GERT)--phase I/II trial: study protocol. BMC Cancer 2006; 6:133. [PMID: 16709245 PMCID: PMC1524973 DOI: 10.1186/1471-2407-6-133] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Accepted: 05/18/2006] [Indexed: 11/12/2022] Open
Abstract
Background The implementation of combined radiochemotherapy (RCHT) with temozolomide (TMZ) has lead to a significant increase in overall survival times in patients with Glioblastoma multiforme (GBM), however, outcome still remains unsatisfactory. The majority of GBMs show an overexpression and/or amplification of the epidermal growth factor receptor (EGFR). Therefore, addition of EGFR-inhibition with cetuximab to the current standard treatment approach with radiotherapy and TMZ seems promising. Methods/design GERT is a one-armed single-center phase I/II trial. In a first step, dose-escalation of TMZ from 50 mg/m2 to 75 mg/m2 together with radiotherapy and cetuximab will be performed. Should safety be proven, the phase II trial will be initiated with the standard dose of 75 mg/m2 of TMZ. Cetuximab will be applied in the standard application dose of 400 mg/m2 in week 1, thereafter at a dose of 250 mg/m2 weekly. A total of 46 patients will be included into this phase I/II trial. Primary endpoints are feasibility and toxicity, secondary endpoints are overall and progression-free survival. An interim analysis will be performed after inclusion of 15 patients into the main study. Patients' enrolment will be performed over a period of 2 years. The observation time will end 2 years after inclusion of the last patient. Discussion The goal of this study is to evaluate the safety and efficacy of combined RCHT-immunotherapy with TMZ and cetuximab as first-line treatment for patients with primary GBM.
Collapse
Affiliation(s)
- Stephanie E Combs
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Steffen Heeger
- Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Renate Haselmann
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Lutz Edler
- Department of Biostatistics, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Daniela Schulz-Ertner
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| |
Collapse
|
15468
|
Yip S, Sabetrasekh R, Sidman RL, Snyder EY. Neural stem cells as novel cancer therapeutic vehicles. Eur J Cancer 2006; 42:1298-308. [PMID: 16697638 DOI: 10.1016/j.ejca.2006.01.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Accepted: 01/23/2006] [Indexed: 01/14/2023]
Abstract
The startling resemblance of many of the behaviours of brain tumours to the intrinsic properties of the neural stem/progenitor cell has triggered a recent dual interest in arming stem cells to track and help eradicate tumours and in viewing stem cell biology as somehow integral to the emergence and/or propagation of the neoplasm itself. These aspects are reviewed and discussed here.
Collapse
Affiliation(s)
- Stephen Yip
- Department of Pathology & Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
15469
|
Reardon DA. Glioblastoma--more questions than answers? ACTA ACUST UNITED AC 2006; 3:60-1. [PMID: 16462825 DOI: 10.1038/ncponc0423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Accepted: 12/01/2005] [Indexed: 11/09/2022]
Affiliation(s)
- David A Reardon
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
15470
|
Magrini SM, Ricardi U, Santoni R, Krengli M, Lupattelli M, Cafaro I, Scoccianti S, Menichelli C, Bertoni F, Enrici RM, Tombolini V, Buglione M, Pirtoli L. Patterns of practice and survival in a retrospective analysis of 1722 adult astrocytoma patients treated between 1985 and 2001 in 12 Italian radiation oncology centers. Int J Radiat Oncol Biol Phys 2006; 65:788-99. [PMID: 16682131 DOI: 10.1016/j.ijrobp.2006.01.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2005] [Revised: 12/31/2005] [Accepted: 01/18/2006] [Indexed: 11/18/2022]
Abstract
PURPOSE To analyze the patterns of practice and survival in a series of 1722 adult astrocytoma patients treated in 12 Italian radiotherapy centers. METHODS AND MATERIALS A total of 1722 patients were treated with postoperative radiotherapy (90% World Health Organization [WHO] Grade 3-4, 62% male, 44% aged >60 years, 25% with severe neurologic deficits, 44% after gross total resection, 52% with high-dose radiotherapy, and 16% with chemotherapy). Variations in the clinical-therapeutic features in three subsequent periods (1985 through 2001) were evaluated, along with overall survival for the different subgroups. RESULTS The proportion of women, of older patients, of those with worse neurologic performance status (NPS), with WHO Grade 4, and with smaller tumors increased with time, as did the proportion of those treated with radical surgery, hypofractionated radiotherapy, and more sophisticated radiotherapy techniques, after staging procedures progressively became more accurate. The main prognostic factors for overall survival were age, sex, neurologic performance status, WHO grade, extent of surgery, and radiation dose. CONCLUSIONS Recently, broader selection criteria for radiotherapy were adopted, together with simpler techniques, smaller total doses, and larger fraction sizes for the worse prognostic categories. Younger, fit patients are treated more aggressively, more often in association with chemotherapy. Survival did not change over time. The accurate evaluation of neurologic status is therefore of utmost importance before the best treatment option for the individual patient is chosen.
Collapse
|
15471
|
Yamanaka R, Arao T, Yajima N, Tsuchiya N, Homma J, Tanaka R, Sano M, Oide A, Sekijima M, Nishio K. Identification of expressed genes characterizing long-term survival in malignant glioma patients. Oncogene 2006; 25:5994-6002. [PMID: 16652150 DOI: 10.1038/sj.onc.1209585] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Better understanding of the underlying biology of malignant gliomas is critical for the development of early detection strategies and new therapeutics. This study aimed to define genes associated with survival. We investigated whether genes coupled with a class prediction model could be used to define subgroups of high-grade gliomas in a more objective manner than standard pathology. RNAs from 29 malignant gliomas were analysed using Agilent microarrays. We identified 21 genes whose expression was most strongly and consistently related to patient survival based on univariate proportional hazards models. In six out of 10 genes, changes in gene expression were validated by quantitative real-time PCR. After adjusting for clinical covariates based on a multivariate analysis, we finally obtained a statistical significance level for DDR1 (discoidin domain receptor family, member 1), DYRK3 (dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 3) and KSP37 (Ksp37 protein). In independent samples, it was confirmed that DDR1 protein expression was also correlated to the prognosis of glioma patients detected by immunohistochemical staining. Furthermore, we analysed the efficacy of the short interfering RNA (siRNA)-mediated inhibition of DDR1 mRNA synthesis in glioma cell lines. Cell proliferation and invasion were significantly suppressed by siRNA against DDR1. Thus, DDR1 can be a novel molecular target of therapy as well as an important predictive marker for survival in patients with glioma. Our method was effective at classifying high-grade gliomas objectively, and provided a more accurate predictor of prognosis than histological grading.
Collapse
Affiliation(s)
- R Yamanaka
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata City, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15472
|
Villano JL, Collins CA, Manasanch EE, Ramaprasad C, van Besien K. Aplastic anaemia in patient with glioblastoma multiforme treated with temozolomide. Lancet Oncol 2006; 7:436-8. [PMID: 16648049 DOI: 10.1016/s1470-2045(06)70696-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- J Lee Villano
- Department of Medicine, Section of Haematology and Oncology, University of Chicago, Chicago, IL, USA.
| | | | | | | | | |
Collapse
|
15473
|
Spence AM, Kiem HP, Partap S, Schuetze S, Silber JR, Peterson RA. Complications of a temozolomide overdose: a case report. J Neurooncol 2006; 80:57-61. [PMID: 16645714 DOI: 10.1007/s11060-006-9152-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Accepted: 03/15/2006] [Indexed: 11/30/2022]
Abstract
PURPOSE AND BACKGROUND This is a report of a 53 year-old man with a glioblastoma multiforme (GBM) treated with an excessive dose of temozolomide (TMZ). METHODS This is a single case review of all clinically relevant records. O6-methylguanine-DNA methyltransferase activity was determined by a biochemical assay. RESULTS Following conventional radiotherapy (RT) without concurrent chemotherapy, the patient received 5,500 mg of TMZ over 2 days. At the standard dose of 200 mg/m2/day his total 5-day dose should have been 1,940 mg. Acutely he had nausea, vomiting and diarrhea for 2 days which cleared. The dominant severe toxicity was pancytopenia between one and four weeks after TMZ which was complicated by secondary infections that were successfully managed. Transient transaminitis occurred but there were no significant pulmonary, renal or other systemic toxicities. His progression free survival was 22 months and overall survival 24 months. CONCLUSION His outcome suggests that TMZ may prove to be a good agent for dose-escalation trials with hematopoietic stem cell rescue.
Collapse
Affiliation(s)
- Alexander M Spence
- Department of Neurology, University of Washington School of Medicine, Room RR650, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| | | | | | | | | | | |
Collapse
|
15474
|
Enting RH, van der Graaf WTA, Kros JM, Heesters M, Metzemaekers J, den Dunnen W. Radiotherapy plus concomitant and adjuvant temozolomide for leptomeningeal pilomyxoid astrocytoma: a case study. J Neurooncol 2006; 80:107-8. [PMID: 16645715 DOI: 10.1007/s11060-006-9151-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Accepted: 03/13/2006] [Indexed: 10/24/2022]
|
15475
|
Abstract
The optimal treatment for patients with oligodendrogliomas is unknown, and current management strategies remain controversial. This past year, further exploration of the molecular genetics of the tumors and its prognostic implications for outcome, evaluation of the utility of positron emission tomography imaging, and the role of radiation and chemotherapy in the treatment of oligodendrogliomas have been reported. It is becoming increasingly apparent that oligodendrogliomas are several distinct diseases on a molecular level, and that key genetic derangements can signify a response to treatment and favorable outcome. The added contributions of recent publications consolidates these emerging impressions. Ultimately, the combination of improved imaging techniques, molecular profiling, and new therapies should result in improved outcome with reduced treatment-related toxicity for patients with newly diagnosed, progressive, and recurrent oligodendrogliomas.
Collapse
Affiliation(s)
- Mark Agulnik
- Princess Margaret Hospital, Department of Medicine, Toronto, ON, Canada
| | | |
Collapse
|
15476
|
Shankar S, Zalutsky MR, Friedman H, Vaidyanathan G. Molecular imaging of alkylguanine-DNA alkyltransferase: further evaluation of radioiodinated derivatives of O6-benzylguanine. Nucl Med Biol 2006; 33:399-407. [PMID: 16631089 DOI: 10.1016/j.nucmedbio.2005.12.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Revised: 12/19/2005] [Accepted: 12/26/2005] [Indexed: 11/17/2022]
Abstract
PURPOSE An inverse correlation has been established between tumor levels of the DNA repair protein alkylguanine-DNA alkyltransferase (AGT) and a positive outcome after alkylator chemotherapy. Quantitative imaging of AGT could provide important information for patient-specific cancer treatment. Several radiolabeled analogues of O6-benzylguanine (BG), a potent AGT inactivator, have been developed and shown to be capable of labeling pure AGT protein. Herein, two of these analogues--O6-3-[*I]iodobenzylguanine ([*I]IBG) and O6-3-[*I]iodobenzyl-2'-deoxyguanosine ([*I]IBdG)--were further evaluated in two murine xenograft models. (AcO)2-[131I]IBdG, a peracetylated derivative of IBdG, also was investigated as an alternative agent. METHODS Several biodistribution studies of radioiodinated IBG and IBdG were performed in TE-671 human rhabdomyosarcoma and DAOY human medulloblastoma murine xenograft models. Mice were treated with BG or its nucleoside analogue dBG to deplete the tumor AGT content. The effect of unlabeled IBG and that of 7,8-benzoflavone (BF), an inhibitor of the cytochrome P-450 isozyme CYP1A2, on the tumor uptake of the tracers was determined. The uptake of (AcO)2-[131I]IBdG along with that of [125I]IBdG in DAOY cells in vitro was determined in the presence and absence of a nucleoside transporter inhibitor, dipyridamole. RESULTS Pretreatment of mice either with BG or dBG failed to reduce tumor levels of [*I]IBG or [*I]IBdG even though such treatments completely depleted tumor AGT content. Treatment of mice with BF increased tumor uptake of [125I]IBG by 56%; however, differentiation of tumors with and without AGT still was not possible. (AcO)2-[131I]IBdG, a peracetylated derivative of IBdG, had a higher uptake in vitro in DAOY tumor cells. However, its uptake, like that of [125I]IBdG, was blocked by dipyridamole. CONCLUSIONS Taken together, these results suggest that labeled agents that are more specific for cellular AGT and that are more metabolically stable are needed.
Collapse
Affiliation(s)
- Sriram Shankar
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
15477
|
Gomez GG, Varella-Garcia M, Kruse CA. Isolation of immunoresistant human glioma cell clones after selection with alloreactive cytotoxic T lymphocytes: cytogenetic and molecular cytogenetic characterization. ACTA ACUST UNITED AC 2006; 165:121-34. [PMID: 16527606 PMCID: PMC1447520 DOI: 10.1016/j.cancergencyto.2005.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Revised: 07/26/2005] [Accepted: 08/01/2005] [Indexed: 01/01/2023]
Abstract
Intratumoral heterogeneity and genetic instability within gliomas may allow intrinsically immunoresistant (IR) cells to escape alloreactive cytotoxic T lymphocyte (aCTL) cellular immunotherapy. The potential existence of aCTL-resistant variants prompted us to investigate whether cellular immunotherapy resistant glioma models could be isolated. To generate the models, repeated intermittent or continuous selective pressure (ISP or CSP) with multiple aCTL populations was applied to a low-passage glioblastoma cell explant, 13-06-MG, obtained from a patient at initial diagnosis. IL-6 and IL-8 secretion was greater in coincubates of aCTL cells with 13-06-ISP and 13-06-CSP immunoselected cells than those with 13-06-MG parental cells. Initially, the immunoselected cells were less sensitive to aCTL lysis; however, the reduced aCTL-sensitivity was not maintained upon further selection. We therefore isolated IR clones from continuously immunoselected cells (13-06-CSP). The frequency of IR clones was 1-6 cells per 10,000 immunoselected cells. Two clones selected for further study, 13-06-IR29 and 13-06-IR30, resisted aCTL lysis in the absence of immunoselective pressure. Cytogenetic analyses revealed structural anomalies and genomic imbalances unique to the IR clones. Based on these findings, a hypothetical model is proposed that traces the origin of the IR clones to a clonal variant within the 13-06-CSP and 13-06-MG populations.
Collapse
Affiliation(s)
- German G. Gomez
- Department of Pathology, University of Colorado Health Sciences Center, Denver, CO 80262
| | | | - Carol A. Kruse
- Division of Cancer Biology and Brain Tumor Research Program, La Jolla Institute for Molecular Medicine, 4570 Executive Boulevard, Suite 100, San Diego, CA 92121
- * Corresponding author. Tel.: (858) 587-8788 ext. 142; fax: (858) 587-6742. E-mail address: (C.A. Kruse)
| |
Collapse
|
15478
|
Blumenthal DT, Schulman SF. Survival outcomes in glioblastoma multiforme, including the impact of adjuvant chemotherapy. Expert Rev Neurother 2006; 5:683-90. [PMID: 16162092 DOI: 10.1586/14737175.5.5.683] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioblastoma is an uncommon cancer, but one that is disproportionately represented in mortality rates. Recent developments in adjuvant chemotherapy have regenerated enthusiasm for the treatment of this tumor. Ongoing translational and clinical research has led to a greater understanding of the biologic and molecular behavior and heterogeneity of this tumor. Recent shifts in treatment standards, as well as further selective individualizing of therapies based on molecular information, promise progress for this difficult-to-treat neoplasm.
Collapse
Affiliation(s)
- Deborah T Blumenthal
- Huntsman Cancer Institute at the University of Utah, 2000 Circle of Hope, Ste 2152, Salt Lake City, UT 84112, USA.
| | | |
Collapse
|
15479
|
Dehdashti AR, Hegi ME, Regli L, Pica A, Stupp R. New trends in the medical management of glioblastoma multiforme: the role of temozolomide chemotherapy. Neurosurg Focus 2006; 20:E6. [PMID: 16709037 DOI: 10.3171/foc.2006.20.4.3] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Standard care for newly diagnosed glioblastoma multiforme (GBM) previously consisted of resection to the greatest extent feasible, followed by radiotherapy. The role of chemotherapy was controversial and its efficacy was marginal at best. Five years ago temozolomide (TMZ) was approved specifically for the treatment of recurrent malignant glioma. The role of TMZ chemotherapy administered alone or as an adjuvant therapy for newly diagnosed GBM has been evaluated in a large randomized trial whose results suggested a significant prolongation of survival following treatment. Findings of correlative molecular studies have indicated that methylguanine methyltransferase promoter methylation may be used as a predictive factor in selecting patients most likely to benefit from such treatment. In this short review the authors summarize the current role of TMZ chemotherapy in the management of GBM, with an emphasis on approved indications and practical aspects.
Collapse
Affiliation(s)
- Amir R Dehdashti
- Department of Neurosurgery, Multidisciplinary Oncology Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
15480
|
Abstract
Following the seminal trial conducted by the European Organisation for Research and Treatment of Cancer (EORTC) and the National Cancer Institute of Canada (NCIC), concurrent temozolomide and radiotherapy has become the new standard of care for patients with newly diagnosed glioblastoma multiforme (GBM). Investigation of emerging therapies (which are now used as salvage therapy) such as small-molecule inhibitors (for example, epidermal growth factor receptor inhibitors) and convection-enhanced delivery (CED) of targeted toxins (for example, interleukin-13/pseudomonas exotoxin) is likely to build on the EORTC/NCIC treatment platform and will, it is hoped, improve survival rates in patients with GBM. The majority of adjuvant Phase I and II trials being conducted by the brain tumor consortia are based on the EORTC/NCIC treatment platform and have added a targeted therapy in an effort to find a promising synergistic treatment. Furthermore, researchers in the consortia are continuing to explore treatments for recurrent GBM, not otherwise eligible for local therapies, such as CED. The treatments under study include novel cytotoxic chemotherapy as well as small-molecule inhibitors; these are being assessed in a variety of Phase I or II trials.
Collapse
Affiliation(s)
- Marc C Chamberlain
- Department of Interdisciplinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612-0804, USA.
| |
Collapse
|
15481
|
Abstract
Unlike high-grade glioma in adults, these tumors represent a minority of all primary central nervous system neoplasms in the pediatric age group (< 22 years). Treatment is quite challenging because of the resistance of high-grade glioma in children to radiotherapy and chemotherapy. Whereas maximum resection and radiotherapy are the mainstay of treatment for this type of tumor in children > 3 years, the role of chemotherapy is less clear. Despite the use of multimodality therapy, less than 20 percent of these children are long-term survivors. This review provides an overview of relevant past, current and future treatment strategies for high-grade glioma in children.
Collapse
Affiliation(s)
- Alberto Broniscer
- Department of Hematology-Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
15482
|
Shapiro WR, Carpenter SP, Roberts K, Shan JS. 131I-chTNT-1/B mAb: tumour necrosis therapy for malignant astrocytic glioma. Expert Opin Biol Ther 2006; 6:539-45. [PMID: 16610983 DOI: 10.1517/14712598.6.5.539] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Treatment of malignant glioma is therapeutically challenging. Despite improvements in neurosurgery, radiotherapy and chemotherapy, few patients diagnosed with anaplastic astrocytoma (AA) or glioblastoma multiforme (GBM) (WHO grades 3 and 4, respectively) will live beyond 2 years. Poor survival is due to the highly invasive nature and protected location of these tumours. Most malignant gliomas cannot be completely resected or irradiated due to their ability to infiltrate diffusely into normal brain tissue. Brain tissue is protected from the systemic circulation via the blood-brain barrier (BBB), which impedes entry of water-soluble chemotherapeutic agents into the tumour at therapeutic concentrations. (131)I-chTNT-1/B mAb (Cotara) employs an innovative strategy to treat the invasive portion of the tumour and the core lesion. (131)I-chTNT-1/B mAb is a genetically engineered, radiolabelled, chimeric monoclonal antibody specific for a universal intracellular antigen (i.e., DNA/histone H1 complex) exposed in the necrotic core of malignant gliomas. This antigen provides an abundant, insoluble, non-diffusible anchor for the mAb. Once localised to necrotic regions of the tumour, (131)I-chTNT-1/B mAb delivers a cytotoxic dose of (131)I radiation to the core lesion. (131)I-chTNT-1/B mAb is delivered via convection-enhanced delivery in order to maximise coverage to the tumour and the invasive front of the glial tumour. The clinical experience to date with (131)I-chTNT-1/B mAb is presented.
Collapse
Affiliation(s)
- William R Shapiro
- Division of Neurology, Barrow Neurological Institute, St. Joseph's Hospital, Phoenix, AZ 85013, USA.
| | | | | | | |
Collapse
|
15483
|
Wang L, Wei Q, Wang LE, Aldape KD, Cao Y, Okcu MF, Hess KR, El-Zein R, Gilbert MR, Woo SY, Prabhu SS, Fuller GN, Bondy ML. Survival prediction in patients with glioblastoma multiforme by human telomerase genetic variation. J Clin Oncol 2006; 24:1627-32. [PMID: 16575014 DOI: 10.1200/jco.2005.04.0402] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most common and aggressive glioma with the poorest survival. Use of biomarkers for screening patients with GBM may be used to modify treatments and improve outcomes. The level of human telomerase (hTERT) expression is an independent predictor of outcome of many cancers, and a functional variant of hTERT MNS16A (shorter tandem repeats or short [S] allele) is associated with increased hTERT mRNA expression. We investigated whether hTERT MNS16A variant genotype predicted survival in GBM patients. PATIENTS AND METHODS We genotyped hTERT MNS16A in 299 GBM patients using polymerase chain reaction and determined hTERT genotype by classifying the DNA band of 243 or 272 base pairs (bp) as S allele and 302 or 333 bp as long (L) allele. We compared overall survival using Kaplan-Meier estimates and equality of survival distributions using the log-rank test, and we computed univariate and multivariate Cox proportional hazards models to estimate the effects of selected variables. RESULTS Overall survival differed significantly by hTERT MNS16A genotype, with median survivals of 25.1, 14.7, and 14.6 months for the SS, SL, and LL genotypes, respectively. Compared with the SS genotype, the hazard ratios for the SL and LL genotypes were 1.69 and 1.87, respectively, after adjustment for other factors. Multivariate Cox regression analysis showed an independent statistically significant association between the hTERT MNS16A variant genotype and outcome. CONCLUSION A functional hTERT MNS16A genotype is a potential biomarker for assessment of survival outcome of GBM. Larger studies are needed to verify these findings.
Collapse
Affiliation(s)
- Luo Wang
- Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15484
|
Henriksson R, Malmström A, Bergström P, Bergh G, Trojanowski T, Andreasson L, Blomquist E, Jonsborg S, Edekling T, Salander P, Brännström T, Bergenheim AT. High-grade astrocytoma treated concomitantly with estramustine and radiotherapy. J Neurooncol 2006; 78:321-6. [PMID: 16598426 DOI: 10.1007/s11060-005-9106-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2005] [Accepted: 12/13/2005] [Indexed: 11/25/2022]
Abstract
Experimental and early clinical investigations have demonstrated encouraging results for estramustine in the treatment of malignant glioma. The present study is an open randomized clinical trial comparing estramustine phosphate (Estracyt) in addition to radiotherapy with radiotherapy alone as first line treatment of astrocytoma grade III and IV. The 140 patients included were in a good clinical condition with a median age of 55 years (range 22-87). Estramustine was given orally, 280 mg twice daily, as soon as the diagnosis was established, during and after the radiotherapy for a period of in total 3 months. Radiotherapy was delivered on weekdays 2 Gy daily up to 56 Gy. Eighteen patients were excluded due to misclassification, leaving 122 patients eligible for evaluation. Overall the treatment was well tolerated. Mild or moderate nausea was the most common side effect of estramustine. The minimum follow-up time was 5.2 years for the surviving patients. For astrocytoma grade III the median survival time was 10.6 (1.3-92.7) months for the radiotherapy only group and 17.3 (0.4-96.9+) months for the estramustine + radiotherapy group. In grade IV the corresponding median survival time was 12.3 (2.1-89.2) and 10.3 (0.3-91.7+) months, respectively. Median time to progress for radiotherapy only and radiotherapy and estramustin group in grade III tumours was 6.5 and 10.1 months, respectively. In grade IV tumours the corresponding figures were 5.1 and 3.3 months, respectively. Although there was a tendency for improved survival in grade III, no statistical significant differences were found between the treatment groups. No differences between the two treatment groups were evident with respect to quality of life according to the EORTC QLQ-protocol. In conclusion, this first randomized study did not demonstrate any significant improvement of using estramustine in addition to conventional radiotherapy, however, a trend for a positive response for the estramustine group was found in patients with grade III glioma.
Collapse
Affiliation(s)
- Roger Henriksson
- Department of Radiation Sciences and Oncology, Umeå University Hospital, Umeå, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15485
|
Moskowitz SI, Jin T, Prayson RA. Role of MIB1 in predicting survival in patients with glioblastomas. J Neurooncol 2006; 76:193-200. [PMID: 16234986 DOI: 10.1007/s11060-005-5262-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Histologic immunomarkers of cell cycle proteins have been utilized for prognosis in high-grade astrocytic tumors. One such marker, MIB1, an antibody immunoreactive throughout the cell cycle, is predictive of more aggressive disease and poorer prognosis in astrocytomas. An independent role of MIB1 analysis for survival prediction and clinical management within histologic grades has not been clearly proven. METHODS This study retrospectively evaluated MIB1 reactivity in tissue samples from 116 patients with glioblastomas on initial medical presentation. Clinical variables considered included gender, age, Karnofsky Performance Scores (KPS), extent of surgical resection, adjuvant radiation and survival. RESULTS Univariate and multivariate analyses were used to correlate these variables with MIB1 staining. MIB1 staining does not predict overall survival or response to adjuvant therapy as an independent risk factor. CONCLUSION MIB1 labeling does not predict patient survival as an independent variable and does not predict response to additional therapies. Patient survival with glioblastoma was predicted by KPS, age, extent of resection and use of adjuvant radiotherapy.
Collapse
Affiliation(s)
- Shaye I Moskowitz
- Department of Neurosurgery, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
15486
|
Vajkoczy P, Knyazev P, Kunkel A, Capelle HH, Behrndt S, von Tengg-Kobligk H, Kiessling F, Eichelsbacher U, Essig M, Read TA, Erber R, Ullrich A. Dominant-negative inhibition of the Axl receptor tyrosine kinase suppresses brain tumor cell growth and invasion and prolongs survival. Proc Natl Acad Sci U S A 2006; 103:5799-804. [PMID: 16585512 PMCID: PMC1458653 DOI: 10.1073/pnas.0510923103] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Malignant gliomas remain incurable brain tumors because of their diffuse-invasive growth. So far, the genetic and molecular events underlying gliomagenesis are poorly understood. In this study, we have identified the receptor tyrosine kinase Axl as a mediator of glioma growth and invasion. We demonstrate that Axl and its ligand Gas6 are overexpressed in human glioma cell lines and that Axl is activated under baseline conditions. Furthermore, Axl is expressed at high levels in human malignant glioma. Inhibition of Axl signaling by overexpression of a dominant-negative receptor mutant (AXL-DN) suppressed experimental gliomagenesis (growth inhibition >85%, P < 0.05) and resulted in long-term survival of mice after intracerebral glioma cell implantation when compared with Axl wild-type (AXL-WT) transfected tumor cells (survival times: AXL-WT, 10 days; AXL-DN, >72 days). A detailed analysis of the distinct hallmarks of glioma pathology, such as cell proliferation, migration, and invasion and tumor angiogenesis, revealed that inhibition of Axl signaling interfered with cell proliferation (inhibition 30% versus AXL-WT), glioma cell migration (inhibition 90% versus mock and AXL-WT, P < 0.05), and invasion (inhibition 62% and 79% versus mock and AXL-WT, respectively; P < 0.05). This study describes the identification, functional manipulation, in vitro and in vivo validation, and preclinical therapeutic inhibition of a target receptor tyrosine kinase mediating glioma growth and invasion. Our findings implicate Axl in gliomagenesis and validate it as a promising target for the development of approaches toward a therapy of these highly aggressive but, as yet, therapy-refractory, tumors.
Collapse
Affiliation(s)
- Peter Vajkoczy
- Department of Neurosurgery, Medical Faculty of the University of Heidelberg, D-68167 Mannheim, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15487
|
Abstract
✓ Malignant gliomas, among which glioblastomas constitute the largest group, are characterized by a dramatically diffuse infiltration into the brain parenchyma with, as a consequence, the fact that no patient with glioblastoma multiforme (GBM) has been cured to date. Migrating GBM cells are resistant to apoptosis (Type I programmed cell death), and thus to radiotherapy and conventional chemotherapy, because of the constitutive activation of several intracellular signaling pathways, of which the most important identified to date are the pathways controlled by phosphatidylinositol 3-kinase, Akt, and the mammalian target of rapamycin (mTOR). Migrating GBM cells seem to be less prone to resist autophagy (Type II programmed cell death), and disruption of the pathway controlled by mTOR induces marked autophagic processes in GBM cells. Temozolomide is the most efficacious cytotoxic drug employed today to combat glioblastoma, and this drug exerts its cytotoxic activity through proautophagic processes. Thus, autophagy represents a kind of Trojan horse that can be used to bypass, at least partly, the dramatic resistance of glioblastoma to radiotherapy and proapoptotic-related chemotherapy.
Collapse
Affiliation(s)
- Florence Lefranc
- Department of Neurosurgery, Erasme University Hospital, Belgium.
| | | |
Collapse
|
15488
|
Hassler M, Seidl S, Fazeny-Doerner B, Preusser M, Hainfellner J, Rössler K, Prayer D, Marosi C. Diversity of cytogenetic and pathohistologic profiles in glioblastoma. ACTA ACUST UNITED AC 2006; 166:46-55. [PMID: 16616111 DOI: 10.1016/j.cancergencyto.2005.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 08/02/2005] [Accepted: 08/22/2005] [Indexed: 11/17/2022]
Abstract
We present a small series of patients with primary glioblastoma multiforme (GBM), and combine individual genetic data with pathohistologic characteristics and clinical outcome. Eighteen patients (12 men, 6 women, median age 51 years) with histologically proven GBM underwent surgical debulking followed by radiotherapy. Fifteen received concomitant chemotherapy. Histologic typing, immunohistochemistry for CD34, karyotypic analysis, and classification of the pattern of neovascularization was done in all patients. In 12/18, we performed methylation-specific polymerase chain reaction of the MGMT gene (O-6-methylguanine-DNA methyltransferase). The survival duration of patients spanned 3-58 months. By classical banding methods, 15/18 patients showed at least one aberration characteristic for primary glioblastoma (+7 in 7/18, deletions of 9p in 10/18 and -10 or deletions from 10q in 8/18 patients). We could not assess whether patients who survived for longer periods showed less complex or fewer aberrations than the patients who survived less than one year. Losses of 6p21(VEGF), 4q27(bFGF), and 12p11 approximately p13 (ING4) were associated with the "bizarre" pattern of neoangiogenesis. Methylation of the MGMT promoter was found in 3/12 patients. Even in this small series, the main characteristic of GBM was its diversity regarding all investigated histologic and genetic characteristics. This extreme diversity should be considered in the design of targeted therapies in GBM.
Collapse
Affiliation(s)
- Marco Hassler
- Department of Internal Medicine I, Clinical Division of Oncology, Medical University Vienna, 6i, Währinger Gürtel 18-20, A-1097 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
15489
|
Kuan CT, Wakiya K, Dowell JM, Herndon JE, Reardon DA, Graner MW, Riggins GJ, Wikstrand CJ, Bigner DD. Glycoprotein nonmetastatic melanoma protein B, a potential molecular therapeutic target in patients with glioblastoma multiforme. Clin Cancer Res 2006; 12:1970-82. [PMID: 16609006 DOI: 10.1158/1078-0432.ccr-05-2797] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE More brain tumor markers are required for prognosis and targeted therapy. We have identified and validated promising molecular therapeutic glioblastoma multiforme (GBM) targets: human transmembrane glycoprotein nonmetastatic melanoma protein B (GPNMB(wt)) and a splice variant form (GPNMB(sv), a 12-amino-acid in-frame insertion in the extracellular domain). EXPERIMENTAL DESIGN We have done genetic and immunohistochemical evaluation of human GBM to determine incidence, distribution, and pattern of localization of GPNMB antigens in brain tumors as well as survival analyses. RESULTS Quantitative real-time PCR on 50 newly diagnosed GBM patient tumor samples indicated that 35 of 50 GBMs (70%) were positive for GPNMB(wt+sv) transcripts and 15 of 50 GBMs (30%) were positive for GPNMB(sv) transcripts. Normal brain samples expressed little or no GPNMB mRNA. We have isolated and characterized an anti-GPNMB polyclonal rabbit antiserum (2640) and two IgG2b monoclonal antibodies (mAb; G11 and U2). The binding affinity constants of the mAbs ranged from 0.27 x 10(8) to 9.6 x 10(8) M(-1) measured by surface plasmon resonance with immobilized GPNMB, or 1.7 to 2.1 x 10(8) M(-1) by Scatchard analyses with cell-expressed GPNMB. Immunohistochemical analysis detected GPNMB in a membranous and cytoplasmic pattern in 52 of 79 GBMs (66%), with focal perivascular reactivity in approximately 27%. Quantitative flow cytometric analysis revealed GPNMB cell surface molecular density of 1.1 x 10(4) to 7.8 x 10(4) molecules per cell, levels sufficient for mAb targeting. Increased GPNMB mRNA levels correlated with elevated GPNMB protein expression in GBM biopsy samples. Univariate and multivariate analyses correlated expression of GPNMB with survival of 39 GBM patients using RNA expression and immunohistochemical data, establishing that patients with relatively high mRNA GPNMB transcript levels (wt+sv and wt), >3-fold over normal brain, as well as positive immunohistochemistry, have a significantly higher risk of death (hazard ratios, 3.0, 2.2, and 2.8, respectively). CONCLUSIONS Increased mRNA and protein levels in GBM patient biopsy samples correlated with higher survival risk; as a detectable surface membrane protein in glioma cells, the data indicate that GPNMB is a potentially useful tumor-associated antigen and prognostic predictor for therapeutic approaches with malignant gliomas or any malignant tumor that expresses GPNMB.
Collapse
Affiliation(s)
- Chien-Tsun Kuan
- Preston Robert Tisch Brain Tumor Center at Duke, Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15490
|
Durando X, Thivat E, Roché H, Bay JO, Lemaire JJ, Verrelle P, Lentz MA, Chazal J, Curé H, Chollet P. Cystemustine in recurrent high grade glioma. J Neurooncol 2006; 79:33-7. [PMID: 16575534 DOI: 10.1007/s11060-005-9096-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Accepted: 12/06/2005] [Indexed: 10/24/2022]
Abstract
In this study, we have assessed the efficacy of a nitrosourea, cystemustine, in treating patients with recurrent high grade glioma with overall survival analysis as primary end-point. Forty-eight patients with recurrent high grade glioma (24 glioblastomas, 17 astrocytomas and 5 oligodendrogliomas) were treated every 2 weeks with 60 mg/m2 cystemustine by a 15 min-infusion. The median number of treatment cycles was 4 (range 1-17). The median overall survival was 8.3 months (range 1-97) and the 6- and 12-month overall survival rates were 55.3% (95% CI, 41.3-68.6%) and 29.8% (95% CI, 18.6-44.0%), respectively. The objective response rate was 18.8% (95% CI, 7.7-29.9%), and 54.2% of patients had stable disease (95% CI, 40.1-68.3%). Multivariate analysis showed that WHO performance status, histology and response to cystemustine were significant prognostic factors for survival of patients with recurrent glioma. In conclusion, cystemustine has encouraging activity for patients with recurrent high grade glioma.
Collapse
Affiliation(s)
- X Durando
- Centre Jean Perrin, 63011, Clermont-Ferrand Cédex, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15491
|
Clavreul A, Delhaye M, Jadaud E, Menei P. Effects of syngeneic cellular vaccinations alone or in combination with GM-CSF on the weakly immunogenic F98 glioma model. J Neurooncol 2006; 79:9-17. [PMID: 16575532 DOI: 10.1007/s11060-005-9115-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Accepted: 12/30/2005] [Indexed: 01/02/2023]
Abstract
Cancer vaccines are one approach for the treatment of brain tumors. Most experimental studies are performed on so-called "immunogenic" brain tumor models such as the rat 9L glioma which does not reflect characteristics of human glioblastoma. In the present study, we tested syngeneic cellular vaccinations alone or in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) on the weakly immunogenic F98 glioma model. Previous studies have shown the efficacy of this treatment on the 9L glioma model. Fisher rats received an intracerebral implantation of F98 cells. Three days later, two subcutaneous vaccinations with irradiated F98 cells were realized in presence or absence of GM-CSF. This scheme of vaccination induced a systemic cellular and humoral immune response capable of in vitro cytolytic activity against F98 cells. However, no significant differences in survival times were noted between vaccinated and untreated animals. Animals vaccinated with GM-CSF or without GM-CSF had respectively a survival time of 26 +/- 2.1 and 25 +/- 4.4 days following tumor challenge versus 26.5 +/- 2.4 days for untreated rats. Fourteen days after the intracerebral tumor implantation, the tumors of vaccinated animals showed a robust infiltration by T lymphocytes, NK cells, dendritic cells, granulocytes and CD11b/c+ myeloid cells. This infiltration was nearly absent in untreated animals except for CD11b/c+ myeloid cells. This study shows that, contrary to the 9L glioma model, the F98 glioma model is resistant to syngeneic cellular vaccinations although a strong peripheral and intratumoral immune response can be induced. These results suggest that the F98 glioma is an attractive model to understand the mechanisms of glioma immunotherapy resistance.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, 49033, Angers Cedex 01, France
| | | | | | | |
Collapse
|
15492
|
Hargrave D, Bartels U, Bouffet E. Diffuse brainstem glioma in children: critical review of clinical trials. Lancet Oncol 2006; 7:241-8. [PMID: 16510333 DOI: 10.1016/s1470-2045(06)70615-5] [Citation(s) in RCA: 451] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Diffuse intrinsic brainstem gliomas constitute 15-20% of all CNS tumours in children, and are the main cause of death in children with brain tumours. Many clinical trials have been done over the past three decades, but survival has remained static. More than 90% of children die within 2 years of diagnosis, and conventional fractionated radiation remains the standard treatment. However, median survival differs substantially between clinical trials, suggesting a survival benefit with some strategies. We appraised the consistency between protocols in terms of eligibility criteria, definition and assessment of response and progression, statistical design, and endpoints. Study designs varied substantially, which could explain the differences in outcome, and no treatment has shown a benefit over conventional radiotherapy. However, consistency between protocols (eg, eligibility criteria and outcome measures) is important to measure the progress in management of diffuse pontine gliomas.
Collapse
Affiliation(s)
- Darren Hargrave
- Department of Paediatric Oncology, Royal Marsden Hospital, Sutton, Surrey, UK
| | | | | |
Collapse
|
15493
|
Trudeau ME, Crump M, Charpentier D, Yelle L, Bordeleau L, Matthews S, Eisenhauer E. Temozolomide in metastatic breast cancer (MBC): a phase II trial of the National Cancer Institute of Canada - Clinical Trials Group (NCIC-CTG). Ann Oncol 2006; 17:952-6. [PMID: 16565212 DOI: 10.1093/annonc/mdl056] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Temozolomide is an oral alkylating agent that crosses the blood-brain barrier, and has preclinical activity in breast cancer. This phase II trial sought to determine the activity and toxicity of temozolomide in metastatic breast cancer (MBC). Temozolomide was administered in a dose dense schedule of 150 mg/m(2) on days 1-7 and 15-21 in a 28-day cycle. MATERIALS AND METHODS Patients had unidimensional disease for response assessment by RECIST criteria, up to two prior chemotherapy regimens for MBC, and may have had brain metastases if radiation was not expected to be required within 4 weeks. RESULTS Nineteen women were entered on the study. All were evaluable for toxicity and 18 were evaluable for response. The median age was 54 years; 14 had prior chemotherapy for MBC and 12 had prior hormones. Sites of disease included bone, brain, liver and lung. Treatment was well tolerated with 14/19 receiving >90% planned dose intensity. Common grade 1-3 drug-related effects included nausea, fatigue, vomiting, anorexia and skin rash. Grade 3-4 hematologic toxicities included granulocytopenia and thrombocytopenia. Of the 18 evaluable patients, there were no objective responses; three had stable disease and 15 progressive disease. CONCLUSIONS No responses to temozolomide were documented in these heavily pretreated women with extensive MBC including brain metastases.
Collapse
Affiliation(s)
- M E Trudeau
- Toronto Sunnybrook Regional Cancer Centre, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
15494
|
Rosenthal MA, Drummond KJ, Dally M, Murphy M, Cher L, Ashley D, Thursfield V, Giles GG. Management of glioma in Victoria (1998–2000): retrospective cohort study. Med J Aust 2006; 184:270-3. [PMID: 16548830 DOI: 10.5694/j.1326-5377.2006.tb00235.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Accepted: 01/05/2006] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To describe the management of and outcomes in a population-based cohort of patients with newly diagnosed glioma. DESIGN, SETTING AND PATIENTS Retrospective cohort study of patients with glioma newly diagnosed over the period 1998-2000 in Victoria. Patients were identified from the population-based Victorian Cancer Registry (VCR). Doctors involved in managing the patients were surveyed by a questionnaire sent out in 2003. The cohort was followed until the end of 2004 to obtain at least 4 years' follow-up data on all patients. MAIN OUTCOME MEASURES Reported treatment, referral patterns and survival rates. RESULTS Over the study period, 992 cases of glioma were identified; 828 completed surveys on eligible patients were obtained (response rate, 93%); 473 patients (57%) had glioblastoma multiforme (GBM); 105 patients (13%) diagnosed with "glioma" had had no histological confirmation. Complete macroscopic resection was performed in 209 patients (25%); 612 patients (74%) were referred for radiotherapy and 326 (54%) for chemotherapy; 39 (5%) were enrolled on a clinical trial. Median survival was 9.2 months for all patients and 7.4 months for patients with GBM. CONCLUSIONS This is the largest reported glioma management survey in the world to date. Much of the patient demographics and approach to treatment were as expected and represent a reasonable "standard of care". However, there are some areas for improvement, including the absence of histological diagnosis in some patients, lack of multidisciplinary care, low clinical trial enrollment and poor use of ancillary services.
Collapse
|
15495
|
Kuijlen JMA, Mooij JJA, Platteel I, Hoving EW, van der Graaf WTA, Span MM, Hollema H, den Dunnen WFA. TRAIL-receptor expression is an independent prognostic factor for survival in patients with a primary glioblastoma multiforme. J Neurooncol 2006; 78:161-71. [PMID: 16544055 DOI: 10.1007/s11060-005-9081-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Accepted: 11/21/2005] [Indexed: 11/29/2022]
Abstract
PURPOSE In order to improve the survival of patients with a glioblastoma multiforme tumor (GBM), new therapeutic strategies must be developed. The use of a death inducing ligand such as TRAIL (TNF Related Apoptosis Inducing Ligand) seems a promising innovative therapy. The aim of this study was to quantify the expression of the death regulating receptors TRAIL-R1, TRAIL-R2 and TRAIL on primary GBM specimens and to correlate this expression with survival. EXPERIMENTAL DESIGN Expression of TRAIL and TRAIL-receptors was assessed by immunohistochemistry, both quantitatively (% of positive tumor cells) and semi-quantitatively (staining intensity) within both the perinecrotic and intermediate tumor zones of primary GBM specimens. RT-PCR of GBM tissue was performed to show expression of TRAIL receptor mRNA. RESULTS Immunohistochemistry showed a slight diffuse intracytoplasmic and a stronger membranous staining for TRAIL and TRAIL receptors in tumor cells. Semi-quantitative expression of TRAIL showed a significantly higher expression of TRAIL in the perinecrotic zone than in the intermediate zone of the tumor (P=0.0001). TRAIL-R2 expression was significantly higher expressed than TRAIL-R1 (P=0.005). The antigenic load of TRAIL-R2 was positively correlated with survival (P=0.02). Multivariate analysis of TRAIL-R1 within the study group (n=62) showed that age, gender, staining intensity, antigenic load, % of TRAIL-R1 expression, were not statistically correlated with survival however radiotherapy was significantly correlated (multivariate analysis: age: P=0.15; gender: P=0.64; staining intensity: P=0.17; antigenic load: P=0.056; % of TRAIL-R1 expression: P=0.058; radiotherapy: P=0.0001). Subgroup analysis of patients who had received radiotherapy (n=47) showed a significant association of % of TRAIL-R1 expression and the antigenic load of TRAIL-R1 with survival (multivariate analysis: P=0.036, respectively, P=0.023). Multivariate analysis of TRAIL-R2 staining intensity and antigenic load, within the study group (P=0.004, respectively, P=0.03) and the subgroup (P=0.002, respectively, P=0.004), showed a significant association with survival. RT-PCR analysis detected a negative relation between the amount of TRAIL-R1 mRNA and the WHO grade of astrocytic tumors (P=0.03). CONCLUSIONS TRAIL-R1 and TRAIL-R2 expression on tumor cells are independent prognostic factors for survival in patients with a glioblastoma multiforme. Both receptors could be targets for TRAIL therapy. As TRAIL-R2 is more expressed, in comparison with TRAIL-R1, on GBM tumor cells, TRAIL-R2 seems to be of more importance as a target for future TRAIL therapy than TRAIL-R1.
Collapse
Affiliation(s)
- Jos M A Kuijlen
- Department of Neurosurgery, University Hospital Groningen, Groningen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
15496
|
Lai R, Chu R, Fraumeni M, Thabane L. Quality of randomized controlled trials reporting in the primary treatment of brain tumors. J Clin Oncol 2006; 24:1136-44. [PMID: 16505433 DOI: 10.1200/jco.2005.03.1179] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To assess the reporting quality of randomized controlled trials (RCTs) in the primary treatment of brain tumors and to identify significant predictors of quality. PATIENTS AND METHODS Two investigators searched MEDLINE, EMBASE, and bibliographies of retrieved articles for RCTs in the primary treatment of brain tumors published between January 1990 and December 2004. We assessed the quality of overall reporting and key methodologic factors reporting (allocation concealment, blinding, and intention to treat [ITT]). Two investigators also rated articles independently using items from the revised Consolidated Standards of Reporting Trials statement. A generalized estimated equation was used to generate regression models that identified significant factors associated with quality of reporting. RESULTS We retrieved 74 relevant RCTs that randomly assigned 14,498 brain tumor patients. The quality of overall reporting has improved during the last 15 years, but eight of the 15 methodologic items were reported in less than 50% of trials. In the appraisal of the reporting quality of key methodologies, allocation concealment, blinding, and adherence to the ITT principle were reported in less than 30% of articles. Multivariable regression models revealed that an impact factor more than 1.66, publication after 1995, and sample size more than 280 were significant factors associated with better overall reporting, whereas complete industrial funding, impact factors more than 2.64, and positive primary outcomes were predictors of higher ratings of the three most important methodologic qualities. CONCLUSION Despite improvement in general reporting quality, key methodologies that safeguard against biases may still benefit from better description. Significant factors associated with better reporting may act as surrogates for other characteristics.
Collapse
Affiliation(s)
- Rose Lai
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.
| | | | | | | |
Collapse
|
15497
|
Donaldson SS, Laningham F, Fisher PG. Advances toward an understanding of brainstem gliomas. J Clin Oncol 2006; 24:1266-72. [PMID: 16525181 DOI: 10.1200/jco.2005.04.6599] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The diagnosis of brainstem glioma was long considered a single entity. However, since the advent of magnetic resonance imaging in the late 1980s, neoplasms within this anatomic region are now recognized to include several tumors of varying behavior and natural history. More recent reports of brainstem tumors include diverse sites such as the cervicomedullary junction, pons, midbrain, or the tectum. Today, these tumors are broadly categorized as either diffuse intrinsic gliomas, most often in the pons, or the nondiffuse brainstem tumors originating at the tectum, focally in the midbrain, dorsal and exophytic to the brainstem, or within the cervicomedullary junction. Although we briefly discuss the nondiffuse tumors, we focus specifically on those diffuse brainstem tumors that regrettably still carry a bleak prognosis.
Collapse
Affiliation(s)
- Sarah S Donaldson
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
15498
|
Butowski NA, Sneed PK, Chang SM. Diagnosis and treatment of recurrent high-grade astrocytoma. J Clin Oncol 2006; 24:1273-80. [PMID: 16525182 DOI: 10.1200/jco.2005.04.7522] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
High-grade gliomas represent a significant source of cancer-related death, and usually recur despite treatment. In this analysis of current brain tumor medicine, we review diagnosis, standard treatment, and emerging therapies for recurrent astrocytomas. Difficulties in interpreting radiographic evidence, especially with regard to differentiating between tumor and necrosis, present a formidable challenge. The most accurate diagnoses come from tissue confirmation of recurrent tumor, but a combination of imaging techniques, such as magnetic resonance spectroscopy imaging, may also be relevant for diagnosis. Repeat resection can prolong life, but repeat irradiation of the brain poses serious risks and results in necrosis of healthy brain tissue; therefore, reirradiation is usually not offered to patients with recurrent tumors. We describe the use of conventional radiotherapy, intensity-modulated radiotherapy, brachytherapy, radiosurgery, and photodynamic therapy for recurrent high-grade glioma. The use of chemotherapy is limited by drug distribution and toxicity, but the development of new drug-delivery techniques such as convection-enhanced delivery, which delivers therapeutic molecules at an effective concentration directly to the brain, may provide a way to reduce systemic exposure to cytotoxic agents. We also discuss targeted therapies designed to inhibit aberrant cell-signaling pathways, as well as new experimental therapies such as immunotherapy. The treatment of this devastating disease has so far been met with limited success, but emerging knowledge of neuroscience and the development of novel therapeutic agents will likely give patients new options and require the neuro-oncology community to redefine clinical trial design and strategy continually.
Collapse
Affiliation(s)
- Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143-0350, USA
| | | | | |
Collapse
|
15499
|
Reardon DA, Rich JN, Friedman HS, Bigner DD. Recent advances in the treatment of malignant astrocytoma. J Clin Oncol 2006; 24:1253-65. [PMID: 16525180 DOI: 10.1200/jco.2005.04.5302] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Malignant gliomas, including the most common subtype, glioblastoma multiforme (GBM), are among the most devastating of neoplasms. Their aggressive infiltration in the CNS typically produces progressive and profound disability--ultimately leading to death in nearly all cases. Improvement in outcome has been elusive despite decades of intensive clinical and laboratory research. Surgery and radiotherapy, the traditional cornerstones of therapy, provide palliative benefit, while the value of chemotherapy has been marginal and controversial. Limited delivery and tumor heterogeneity are two fundamental factors that have critically hindered therapeutic progress. A novel chemoradiotherapy approach, consisting of temozolomide administered concurrently during radiotherapy followed by adjuvant systemic temozolomide, has recently demonstrated a meaningful, albeit modest, improvement in overall survival for newly diagnosed GBM patients. As cell-signaling alterations linked to the development and progression of gliomas are being increasingly elucidated, targeted therapies have rapidly entered preclinical and clinical evaluation. Responses to therapies that function via DNA damage have been associated with specific mediators of resistance that may also be subject to targeted therapies. Other approaches include novel locoregional delivery techniques to overcome barriers of delivery. The simultaneous development of multiple advanced therapies based on specific tumor biology may finally offer glioma patients improved survival.
Collapse
Affiliation(s)
- David A Reardon
- Preston Robert Tisch Brain Tumor Center at Duke University, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | |
Collapse
|
15500
|
Abstract
Diffusely infiltrating low-grade gliomas (LGGs) include astrocytomas, oligodendrogliomas, and mixed oligoastrocytomas (WHO grade 2). Due to the routine use of magnetic resonance imaging, there is an increasing need to formulate treatment guidelines for patients with LGGs. However, there is little consensus about the optimal treatment strategy for diffusely infiltrative LGGs, and the clinical management of LGGs is one of the most controversial areas in neurooncology. Although the standard of care has not been established, several randomized trials are beginning to provide some answers. Furthermore, laboratory correlative studies are defining subsets of LGG that may identify patients with better prognoses and increased chance of responding to therapy. This article reviews the most recent data regarding the treatment of LGG, emphasizing evidenced based approaches from current clinical trials.
Collapse
Affiliation(s)
- Frederick F Lang
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|