101
|
Wang Y, Yang J, Wild AT, Wu WH, Shah R, Danussi C, Riggins GJ, Kannan K, Sulman EP, Chan TA, Huse JT. G-quadruplex DNA drives genomic instability and represents a targetable molecular abnormality in ATRX-deficient malignant glioma. Nat Commun 2019; 10:943. [PMID: 30808951 PMCID: PMC6391399 DOI: 10.1038/s41467-019-08905-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 02/08/2019] [Indexed: 12/11/2022] Open
Abstract
Mutational inactivation of ATRX (α-thalassemia mental retardation X-linked) represents a defining molecular alteration in large subsets of malignant glioma. Yet the pathogenic consequences of ATRX deficiency remain unclear, as do tractable mechanisms for its therapeutic targeting. Here we report that ATRX loss in isogenic glioma model systems induces replication stress and DNA damage by way of G-quadruplex (G4) DNA secondary structure. Moreover, these effects are associated with the acquisition of disease-relevant copy number alterations over time. We then demonstrate, both in vitro and in vivo, that ATRX deficiency selectively enhances DNA damage and cell death following chemical G4 stabilization. Finally, we show that G4 stabilization synergizes with other DNA-damaging therapies, including ionizing radiation, in the ATRX-deficient context. Our findings reveal novel pathogenic mechanisms driven by ATRX deficiency in glioma, while also pointing to tangible strategies for drug development. ATRX deficiency is linked to genomic stability in cancer cells. Here, the authors show that ATRX inactivation induces G-quadruplex formation, leading to genome-wide DNA damage, and the use of G-quadruplex stabilisers can be exploited therapeutically in ATRX deficient gliomas.
Collapse
Affiliation(s)
- Yuxiang Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Jie Yang
- Department of Radation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aaron T Wild
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Wei H Wu
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Rachna Shah
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Carla Danussi
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gregory J Riggins
- Departments of Neurosurgery, Oncology, and Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Kasthuri Kannan
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Erik P Sulman
- Department of Radation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Jason T Huse
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
102
|
Herrlinger U, Tzaridis T, Mack F, Steinbach JP, Schlegel U, Sabel M, Hau P, Kortmann RD, Krex D, Grauer O, Goldbrunner R, Schnell O, Bähr O, Uhl M, Seidel C, Tabatabai G, Kowalski T, Ringel F, Schmidt-Graf F, Suchorska B, Brehmer S, Weyerbrock A, Renovanz M, Bullinger L, Galldiks N, Vajkoczy P, Misch M, Vatter H, Stuplich M, Schäfer N, Kebir S, Weller J, Schaub C, Stummer W, Tonn JC, Simon M, Keil VC, Nelles M, Urbach H, Coenen M, Wick W, Weller M, Fimmers R, Schmid M, Hattingen E, Pietsch T, Coch C, Glas M. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet 2019; 393:678-688. [PMID: 30782343 DOI: 10.1016/s0140-6736(18)31791-4] [Citation(s) in RCA: 370] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/12/2018] [Accepted: 07/27/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND There is an urgent need for more effective therapies for glioblastoma. Data from a previous unrandomised phase 2 trial suggested that lomustine-temozolomide plus radiotherapy might be superior to temozolomide chemoradiotherapy in newly diagnosed glioblastoma with methylation of the MGMT promoter. In the CeTeG/NOA-09 trial, we aimed to further investigate the effect of lomustine-temozolomide therapy in the setting of a randomised phase 3 trial. METHODS In this open-label, randomised, phase 3 trial, we enrolled patients from 17 German university hospitals who were aged 18-70 years, with newly diagnosed glioblastoma with methylated MGMT promoter, and a Karnofsky Performance Score of 70% and higher. Patients were randomly assigned (1:1) with a predefined SAS-generated randomisation list to standard temozolomide chemoradiotherapy (75 mg/m2 per day concomitant to radiotherapy [59-60 Gy] followed by six courses of temozolomide 150-200 mg/m2 per day on the first 5 days of the 4-week course) or to up to six courses of lomustine (100 mg/m2 on day 1) plus temozolomide (100-200 mg/m2 per day on days 2-6 of the 6-week course) in addition to radiotherapy (59-60 Gy). Because of the different schedules, patients and physicians were not masked to treatment groups. The primary endpoint was overall survival in the modified intention-to-treat population, comprising all randomly assigned patients who started their allocated chemotherapy. The prespecified test for overall survival differences was a log-rank test stratified for centre and recursive partitioning analysis class. The trial is registered with ClinicalTrials.gov, number NCT01149109. FINDINGS Between June 17, 2011, and April 8, 2014, 141 patients were randomly assigned to the treatment groups; 129 patients (63 in the temozolomide and 66 in the lomustine-temozolomide group) constituted the modified intention-to-treat population. Median overall survival was improved from 31·4 months (95% CI 27·7-47·1) with temozolomide to 48·1 months (32·6 months-not assessable) with lomustine-temozolomide (hazard ratio [HR] 0·60, 95% CI 0·35-1·03; p=0·0492 for log-rank analysis). A significant overall survival difference between groups was also found in a secondary analysis of the intention-to-treat population (n=141, HR 0·60, 95% CI 0·35-1·03; p=0·0432 for log-rank analysis). Adverse events of grade 3 or higher were observed in 32 (51%) of 63 patients in the temozolomide group and 39 (59%) of 66 patients in the lomustine-temozolomide group. There were no treatment-related deaths. INTERPRETATION Our results suggest that lomustine-temozolomide chemotherapy might improve survival compared with temozolomide standard therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter. The findings should be interpreted with caution, owing to the small size of the trial. FUNDING German Federal Ministry of Education and Research.
Collapse
Affiliation(s)
- Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany.
| | - Theophilos Tzaridis
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Frederic Mack
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | | | - Uwe Schlegel
- Department of Neurology, University Hospital Knappschaftskrankenhaus, Ruhr-Universität Bochum, Bochum, Germany
| | - Michael Sabel
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander Neurooncology Unit, University Hospital Regensburg, Regensburg, Germany
| | | | - Dietmar Krex
- Department of Neurosurgery, University of Dresden, Dresden, Germany
| | - Oliver Grauer
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Oliver Schnell
- Department of Neurosurgery, Ludwig Maximillian University of Munich and German Cancer Consortium, Partner Site Munich, Munich, Germany; Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Oliver Bähr
- Dr Senckenberg Institute of Neurooncology, University of Frankfurt, Frankfurt, Germany
| | - Martin Uhl
- Department of Neurology and Wilhelm Sander Neurooncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Clemens Seidel
- Department of Radiation Oncology, University of Leipzig, Leipzig, Germany
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neurooncology, University of Tübingen, Tübingen, Germany
| | - Thomas Kowalski
- Department of Neurology, University Hospital Knappschaftskrankenhaus, Ruhr-Universität Bochum, Bochum, Germany
| | - Florian Ringel
- Department of Neurosurgery, Technical University of Munich, Munich, Germany; Department of Neurosurgery, University of Mainz, Mainz, Germany
| | | | - Bogdana Suchorska
- Department of Neurosurgery, Ludwig Maximillian University of Munich and German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Stefanie Brehmer
- Department of Neurosurgery, University of Mannheim, Mannheim, Germany
| | - Astrid Weyerbrock
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Miriam Renovanz
- Department of Neurosurgery, University of Mainz, Mainz, Germany
| | - Lars Bullinger
- Department of Internal Medicine, University of Ulm, Ulm, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine (INM-3), Juelich, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité University of Berlin, Berlin, Germany
| | - Martin Misch
- Department of Neurosurgery, Charité University of Berlin, Berlin, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Moritz Stuplich
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Niklas Schäfer
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Sied Kebir
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Johannes Weller
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Christina Schaub
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Walter Stummer
- Department of Neurosurgery, University of Münster, Münster, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Ludwig Maximillian University of Munich and German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Matthias Simon
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Vera C Keil
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Michael Nelles
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Horst Urbach
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany; Department of Neuroradiology, University of Freiburg, Freiburg, Germany
| | - Martin Coenen
- Study Centre Bonn, University Hospital Bonn, Bonn, Germany
| | - Wolfgang Wick
- Department of Neurology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Rolf Fimmers
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Matthias Schmid
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Elke Hattingen
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Torsten Pietsch
- Institute of Neuropathology and DGNN Brain Tumor Reference Centre, University Hospital Bonn, Bonn, Germany
| | - Christoph Coch
- Study Centre Bonn, University Hospital Bonn, Bonn, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany; Division of Clinical Neurooncology, Department of Neurology and West German Cancer Center, German Cancer Consortium, Partner Site Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
103
|
Hohmann T, Feese K, Ghadban C, Dehghani F, Grabiec U. On the influence of cannabinoids on cell morphology and motility of glioblastoma cells. PLoS One 2019; 14:e0212037. [PMID: 30753211 PMCID: PMC6372232 DOI: 10.1371/journal.pone.0212037] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/25/2019] [Indexed: 12/20/2022] Open
Abstract
The mechanisms behind the anti-tumoral effects of cannabinoids by impacting the migratory activity of tumor cells are only partially understood. Previous studies demonstrated that cannabinoids altered the organization of the actin cytoskeleton in various cell types. As actin is one of the main contributors to cell motility and is postulated to be linked to tumor invasion, we tested the following hypothesizes: 1) Can cannabinoids alter cell motility in a cannabinoid receptor dependent manner? 2) Are these alterations associated with reorganizations in the actin cytoskeleton? 3) If so, what are the underlying molecular mechanisms? Three different glioblastoma cell lines were treated with specific cannabinoid receptor 1 and 2 agonists and antagonists. Afterwards, we measured changes in cell motility using live cell imaging and alterations of the actin structure in fixed cells. Additionally, the protein amount of phosphorylated p44/42 mitogen-activated protein kinase (MAPK), focal adhesion kinases (FAK) and phosphorylated FAK (pFAK) over time were measured. Cannabinoids induced changes in cell motility, morphology and actin organization in a receptor and cell line dependent manner. No significant changes were observed in the analyzed signaling molecules. Cannabinoids can principally induce changes in the actin cytoskeleton and motility of glioblastoma cell lines. Additionally, single cell motility of glioblastoma is independent of their morphology. Furthermore, the observed effects seem to be independent of p44/42 MAPK and pFAK pathways.
Collapse
Affiliation(s)
- Tim Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Kerstin Feese
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chalid Ghadban
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Urszula Grabiec
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
104
|
Tabet A, Jensen MP, Parkins CC, Patil PG, Watts C, Scherman OA. Designing Next-Generation Local Drug Delivery Vehicles for Glioblastoma Adjuvant Chemotherapy: Lessons from the Clinic. Adv Healthc Mater 2019; 8:e1801391. [PMID: 30632715 DOI: 10.1002/adhm.201801391] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/03/2018] [Indexed: 12/11/2022]
Abstract
To date, the clinical outcomes and survival rates for patients with glioblastoma (GB) remain poor. A promising approach to disease-modification involves local delivery of adjuvant chemotherapy into the resection cavity, thus circumventing the restrictions imposed by the blood-brain barrier. The clinical performance of the only FDA-approved local therapy for GB [carmustine (BCNU)-loaded polyanhydride wafers], however, has been disappointing. There is an unmet medical need in the local treatment of GB for drug delivery vehicles that provide sustained local release of small molecules and combination drugs over several months. Herein, key quantitative lessons from the use of local and systemic adjuvant chemotherapy for GB in the clinic are outlined, and it is discussed how these can inform the development of next-generation therapies. Several recent approaches are highlighted, and it is proposed that long-lasting soft materials can capture the value of stiff BCNU-loaded wafers while addressing a number of unmet medical needs. Finally, it is suggested that improved communication between materials scientists, biomedical scientists, and clinicians may facilitate translation of these materials into the clinic and ultimately lead to improved clinical outcomes.
Collapse
Affiliation(s)
- Anthony Tabet
- Melville Laboratory for Polymer Synthesis; Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Melanie P. Jensen
- Division of Neurosurgery; Department of Clinical Neurosciences; Addenbrooke's Hospital; University of Cambridge; Hills Road Cambridge CB2 0QQ UK
| | - Christopher C. Parkins
- Melville Laboratory for Polymer Synthesis; Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Parag G. Patil
- Department of Neurosurgery; University of Michigan Medical School; Ann Arbor MI 48109 USA
| | - Colin Watts
- Division of Neurosurgery; Department of Clinical Neurosciences; Addenbrooke's Hospital; University of Cambridge; Hills Road Cambridge CB2 0QQ UK
- Department of Neurosurgery; Birmingham Brain Cancer Program; Institute of Cancer and Genomic Sciences; University of Birmingham; Birmingham B15 2TT UK
| | - Oren A. Scherman
- Melville Laboratory for Polymer Synthesis; Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| |
Collapse
|
105
|
Sadeghi Y, Tabatabaei Irani P, Rafiee L, Tajadini M, Haghjooy Javanmard S. Evaluation of rs1957106 Polymorphism of NF-κBI in Glioblastoma Multiforme in Isfahan, Iran. Adv Biomed Res 2019; 8:9. [PMID: 30820430 PMCID: PMC6385670 DOI: 10.4103/abr.abr_227_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The kB family of nuclear factor (NF-κB) is a series of transcription factors that plays a key role in regulation of immunity, cell growth, and apoptosis and is considered as the main downstream component of epidermal growth factor receptor for which there are evidence of excessive activity in most cases of glioblastoma multiform (GBM). Thus, the current information has gained evidence on NF-κBIA tumor suppressor role in GMB. SNP rs1957106 was diagnosed as a new polymorphism which affected the expression of NF-κBI and causes activation of NF-κB in GBM patients. MATERIALS AND METHODS This study was conducted on 100 cases of GBM including 47 paraffin-embedded brain tissue samples and 53 blood samples from another 53 GBM patients and 150 controls. The NF-κBI rs1957106 SNP was identified by the NCBI, and genotyping was performed by high-resolution melt (HRM) assay. Melt curves from HRM which suspected to single-nucleotide polymorphism (SNP) were selected and subjected to direct sequencing. RESULTS The distribution of allele A of NF-κβ gene in patients with GBM with 31% was not significantly different from healthy participants (27.3%) (P = 0.375). Furthermore, the distribution of AG and GG genotypes in comparison with AA genotypes did not show a significant correlation with GBM incidence (P > 0.05). CONCLUSION Findings of the present study provide evidence that the rs1957106 SNP in NF-κBIA is found more in GBM patients, but it was not statistically significant. As there are conflicting studies showing significant higher rate of this SNP in GBM, further study is suggested.
Collapse
Affiliation(s)
- Yasaman Sadeghi
- From the Department of General Medicine, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pouya Tabatabaei Irani
- From the Department of General Medicine, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Laleh Rafiee
- Department of Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohamadhasan Tajadini
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Department of Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
106
|
A DFT study of the degradation mechanism of anticancer drug carmustine in an aqueous medium. Struct Chem 2019. [DOI: 10.1007/s11224-019-1285-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
107
|
Repolarization of myeloid derived suppressor cells via magnetic nanoparticles to promote radiotherapy for glioma treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 16:126-137. [PMID: 30553919 DOI: 10.1016/j.nano.2018.11.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
Abstract
Although radiotherapy has been established as a major therapeutic modality for glioma, radical new avenues are critically needed to prevent inevitable tumor recurrence. Herein, we utilized a magnetic nanoparticle-based platform with cationic polymer modification to promote radiotherapy for glioma treatment. We found that the nanoplatform induced cytotoxicity to glioma cells under radiation as well as promoting significant survival benefits in both immunocompetent and aythmic mice with glioma. Utilizing the magnetic properties of the nanoparticles, we were able to ascertain that myeloid derived suppressor cells (MDSC) were taking up nanoparticles in the brain tumor. The observed efficacy was attributed to destruction of glioma cells as well as MDSCs repolarization from immunosuppressive phenotype to a pro-inflammatory phenotype, which promoted antitumor effects and synergistically promoted radio-therapeutic effects. Our nanoparticles provide a robust dual-targeting platform for glioma radiotherapy by simultaneous eradication of tumor cells and manipulation of myeloid phenotypes in the central nervous system.
Collapse
|
108
|
Wank M, Schilling D, Schmid TE, Meyer B, Gempt J, Barz M, Schlegel J, Liesche F, Kessel KA, Wiestler B, Bette S, Zimmer C, Combs SE. Human Glioma Migration and Infiltration Properties as a Target for Personalized Radiation Medicine. Cancers (Basel) 2018; 10:cancers10110456. [PMID: 30463322 PMCID: PMC6266328 DOI: 10.3390/cancers10110456] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 01/28/2023] Open
Abstract
Gliomas are primary brain tumors that present the majority of malignant adult brain tumors. Gliomas are subdivided into low- and high-grade tumors. Despite extensive research in recent years, the prognosis of malignant glioma patients remains poor. This is caused by naturally highly infiltrative capacities as well as high levels of radio- and chemoresistance. Additionally, it was shown that low linear energy transfer (LET) irradiation enhances migration and invasion of several glioma entities which might counteract today’s treatment concepts. However, this finding is discussed controversially. In the era of personalized medicine, this controversial data might be attributed to the patient-specific heterogeneity that ultimately could be used for treatment. Thus, current developments in glioma therapy should be seen in the context of intrinsic and radiation-enhanced migration and invasion. Due to the natural heterogeneity of glioma cells and different radiation responses, a personalized radiation treatment concept is suggested and alternative radiation concepts are discussed.
Collapse
Affiliation(s)
- Michaela Wank
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany.
| | - Daniela Schilling
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Thomas E Schmid
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Bernhard Meyer
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Jens Gempt
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Melanie Barz
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Jürgen Schlegel
- Department of Neuropathology, Technical University of Munich (TUM), 81675 Munich, Germany.
| | - Friederike Liesche
- Department of Neuropathology, Technical University of Munich (TUM), 81675 Munich, Germany.
| | - Kerstin A Kessel
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany.
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Stefanie Bette
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Stephanie E Combs
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany.
| |
Collapse
|
109
|
Abstract
Clinical research in neuro-oncology frequently classifies patients over 60-70 years of age as 'elderly', a designation intended to identify patients with the disease characteristics, psychosocial changes, and susceptibility to treatment toxicities associated with advancing age. The elderly account for a large proportion of patients diagnosed with glioblastoma (GBM), and this population is projected to increase. Their prognosis is inferior to that of GBM patients as a whole, and concerns over treatment toxicity may limit the aggressiveness with which they are treated. Recent clinical studies have assisted with therapeutic decision making in this cohort. Hypofractionated radiation with concurrent and adjuvant temozolomide has been shown to increase survival without worsened quality of life in elderly patients with good functional status. Single modality radiation therapy or temozolomide therapy are frequently used in this population, and while neither has demonstrated superiority, O6-methylguanine-DNA methyltransferase (MGMT) methylation status is predictive of improved survival with temozolomide over radiation therapy. Despite these advances, ambiguity as to how to best define, assess, and treat this population remains. The specific response of elderly patients to emerging therapies, such as immunotherapies, is unclear. Advancing outcomes for elderly patients with GBM requires persistent efforts to include them in translational and clinical research endeavors, and concurrent dedication to the preservation of function and quality of life in this population.
Collapse
Affiliation(s)
- Rebecca A Harrison
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX, 77030, USA.
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX, 77030, USA
| |
Collapse
|
110
|
Wilson KM, Mathews-Griner LA, Williamson T, Guha R, Chen L, Shinn P, McKnight C, Michael S, Klumpp-Thomas C, Binder ZA, Ferrer M, Gallia GL, Thomas CJ, Riggins GJ. Mutation Profiles in Glioblastoma 3D Oncospheres Modulate Drug Efficacy. SLAS Technol 2018; 24:28-40. [PMID: 30289729 DOI: 10.1177/2472630318803749] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is a lethal brain cancer with a median survival time of approximately 15 months following treatment. Common in vitro GBM models for drug screening are adherent and do not recapitulate the features of human GBM in vivo. Here we report the genomic characterization of nine patient-derived, spheroid GBM cell lines that recapitulate human GBM characteristics in orthotopic xenograft models. Genomic sequencing revealed that the spheroid lines contain alterations in GBM driver genes such as PTEN, CDKN2A, and NF1. Two spheroid cell lines, JHH-136 and JHH-520, were utilized in a high-throughput drug screen for cell viability using a 1912-member compound library. Drug mechanisms that were cytotoxic in both cell lines were Hsp90 and proteasome inhibitors. JHH-136 was uniquely sensitive to topoisomerase 1 inhibitors, while JHH-520 was uniquely sensitive to Mek inhibitors. Drug combination screening revealed that PI3 kinase inhibitors combined with Mek or proteasome inhibitors were synergistic. However, animal studies to test these drug combinations in vivo revealed that Mek inhibition alone was superior to the combination treatments. These data show that these GBM spheroid lines are amenable to high-throughput drug screening and that this dataset may deliver promising therapeutic leads for future GBM preclinical studies.
Collapse
Affiliation(s)
- Kelli M Wilson
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Lesley A Mathews-Griner
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Tara Williamson
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Rajarshi Guha
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Lu Chen
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Paul Shinn
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Crystal McKnight
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Sam Michael
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Carleen Klumpp-Thomas
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Zev A Binder
- 3 Department of Neurosurgery, Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Ferrer
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Gary L Gallia
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Craig J Thomas
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Gregory J Riggins
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| |
Collapse
|
111
|
Lai X, Deng Z, Guo H, Zhu X, Tu W. HP1α is highly expressed in glioma cells and facilitates cell proliferation and survival. Cancer Biomark 2018; 20:453-460. [DOI: 10.3233/cbm-170249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
112
|
Park T, Min KA, Cheong H, Moon C, Shin MC. Genetic engineering and characterisation of chlorotoxin-fused gelonin for enhanced glioblastoma therapy. J Drug Target 2018; 27:950-958. [DOI: 10.1080/1061186x.2018.1516221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Taehoon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, Republic of Korea
| | - Heesun Cheong
- Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi-do, Republic of Korea
| | - Cheol Moon
- College of Pharmacy, Sunchon National University, Suncheon, Jeonnam, Republic of Korea
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| |
Collapse
|
113
|
The Safety of Bevacizumab Administered Shortly after Laser Interstitial Thermal Therapy in Glioblastoma: A Case Series. World Neurosurg 2018; 117:e588-e594. [DOI: 10.1016/j.wneu.2018.06.092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 11/18/2022]
|
114
|
Abstract
PURPOSE OF REVIEW This article reviews the standard treatment for high-grade gliomas, with a focus on promising new strategies and response assessment. RECENT FINDINGS The new World Health Organization (WHO) classification of central nervous system tumors classifies high-grade gliomas based on molecular markers that are of prognostic and therapeutic significance. The addition of chemotherapy, specifically procarbazine, CCNU (lomustine), and vincristine, to radiation in newly diagnosed 1p/19q codeleted anaplastic oligodendrogliomas doubled overall survival. The US Food and Drug Administration (FDA) recently approved the addition of tumor treating fields to adjuvant temozolomide after radiation with concurrent temozolomide in newly diagnosed glioblastoma. A phase3 trial for recurrent glioblastoma did not show an overall survival benefit for the addition of bevacizumab to lomustine compared to lomustine alone. Current efforts are focused on the development of novel treatment approaches, including molecular targeted agents and immunotherapies. SUMMARY Surgery, radiation, and chemotherapy remain the standard treatment options for patients with high-grade gliomas. Despite aggressive treatment, these tumors progress, and overall outcomes have not changed much in the past decade. However, our understanding of the disease is improving, and newer therapies appear promising.
Collapse
|
115
|
Wick W, Osswald M, Wick A, Winkler F. Treatment of glioblastoma in adults. Ther Adv Neurol Disord 2018; 11:1756286418790452. [PMID: 30083233 PMCID: PMC6071154 DOI: 10.1177/1756286418790452] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/24/2018] [Indexed: 01/25/2023] Open
Abstract
The diagnosis of a glioblastoma is mainly made on the basis of their microscopic appearance with the additional determination of epigenetic as well as mutational analyses as deemed appropriate and taken into account in different centers. How far the recent discovery of tumor networks will stimulate novel treatments is a subject of intensive research. A tissue diagnosis is the mainstay. Regardless of age, patients should undergo a maximal safe resection. Magnetic resonance imaging is the surrogate parameter of choice for follow up. Patients should receive chemoradiotherapy with temozolomide with the radiation schedule adapted to performance status, age and tumor location. The use of temozolomide may be reconsidered according to methylguanine DNA methyltransferase (MGMT) promoter methylation status; patients with an active promoter may be subjected to a trial or further molecular work-up in order to potentially replace temozolomide; patients with an inactive (hypermethylated) MGMT promoter may be counseled for the co-treatment with the methylating and alkylating compound lomustine in addition to temozolomide. Tumor-treating fields are an additive option independent of the MGMT status. Determination of recurrence is still challenging. Patients with clinical or radiographic confirmed progression should be counseled for a second surgical intervention, that is, to reach another macroscopic removal of the tumor bulk or to obtain tissue for an updated molecular analysis. Immune therapeutic approaches may be dependent on tumor types and molecular signatures. In newly diagnosed and recurrent glioblastoma, bevacizumab prolongs progression-free survival without affecting overall survival in an unselected population of glioblastoma patients. Whether or not selection can be made on the basis of molecular or imaging parameters remains to be determined. Some patients may benefit from a second radiotherapy. In our view, the near future will provide support for translating the amazing progress in understanding the molecular background of glioblastoma in to more complex, but promising therapy concepts.
Collapse
Affiliation(s)
- Wolfgang Wick
- Neurology Clinic & National Center for Tumor
Disease, University of Heidelberg, Im Neuenheimer Feld 400, D-69120
Heidelberg, Germany
| | - Matthias Osswald
- Neurology Clinic, University of Heidelberg,
Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antje Wick
- Neurology Clinic, University of Heidelberg,
German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ),
Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic, University of Heidelberg,
Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
116
|
Brain tumor related-epilepsy. Neurol Neurochir Pol 2018; 52:436-447. [PMID: 30122210 DOI: 10.1016/j.pjnns.2018.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Gliomas are commonly associated with the development of epilepsy; in some cases the two conditions share common pathogenic mechanisms and may influence each other. Brain tumor related-epilepsy (BTRE) complicates the clinical management of gliomas and can substantially affect daily life. STATE OF THE ART The incidence of seizures is high in patients with slow growing tumors located in the frontotemporal regions. However, recent studies suggest that epileptogenesis may be more associated with tumor molecular genetic markers than tumor grade or location. Although the exact mechanism of epileptogenesis in glioma is incompletely understood, glutamate-induced excitotoxicity and disruption of intracellular communication have garnered the most attention. CLINICAL MANAGEMENT Management of BTRE requires a multidisciplinary approach involving the use of antiepileptic drugs (AEDs), surgery aided by electrocorticography, and adjuvant chemoradiation. FUTURE DIRECTIONS Insight into the mechanisms of glioma growth and epileptogenesis is essential to identify new treatment targets and to develop effective treatment for both conditions. Selecting AEDs tailored to act against known tumor molecular markers involved in the epileptogenesis could enhance treatment value and help inform individualized medicine in BRTE.
Collapse
|
117
|
Yamamoto Y, Sasaki N, Kumagai K, Takeuchi S, Toyooka T, Otani N, Wada K, Narita Y, Ichimura K, Namba H, Mori K, Tomiyama A. Involvement of Intracellular Cholesterol in Temozolomide-Induced Glioblastoma Cell Death. Neurol Med Chir (Tokyo) 2018; 58:296-302. [PMID: 29899179 PMCID: PMC6048351 DOI: 10.2176/nmc.ra.2018-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) still carries a poor prognosis due to the refractoriness against antitumor drugs. Temozolomide (TMZ), one of the few standard therapy drugs against GBM worldwide, has only limited effect due to acquired TMZ resistance of GBM. Therefore, development of novel therapeutic methods to overcome the TMZ resistance of GBM is urgent. The brain is the most cholesterol-rich organ in the human body, so modulation of cholesterol in tumor cells originating from the brain including GBM may be a tumor-specific therapeutic strategy including enhancement of TMZ effects. The unique lipid metabolism of glioma has recently been reported, but the involvement of intracellular cholesterol in TMZ therapy is yet to be fully elucidated. This review summarizes the effect of modulation of intracellular cholesterol level on cancer therapy including GBM treatment and the implications for TMZ therapy. Our recent findings about the involvement of intracellular cholesterol in TMZ-induced GBM cell death are described.
Collapse
Affiliation(s)
- Yutaro Yamamoto
- Department of Neurosurgery, National Defense Medical College.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | - Nobuyoshi Sasaki
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute.,Department of Neurosurgery, Kyorin University School of Medicine
| | - Kosuke Kumagai
- Department of Neurosurgery, National Defense Medical College
| | - Satoru Takeuchi
- Department of Neurosurgery, National Defense Medical College
| | | | - Naoki Otani
- Department of Neurosurgery, National Defense Medical College
| | - Kojiro Wada
- Department of Neurosurgery, National Defense Medical College
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine
| | - Kentaro Mori
- Department of Neurosurgery, National Defense Medical College
| | - Arata Tomiyama
- Department of Neurosurgery, National Defense Medical College.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| |
Collapse
|
118
|
Using Smaller-Than-Standard Radiation Treatment Margins Does Not Change Survival Outcomes in Patients with High-Grade Gliomas. Pract Radiat Oncol 2018; 9:16-23. [PMID: 30195927 DOI: 10.1016/j.prro.2018.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/10/2018] [Accepted: 06/04/2018] [Indexed: 01/16/2023]
Abstract
PURPOSE The number of studies that evaluate treatment margins for high grade gliomas (HGG) are limited. We hypothesize that patients with HGG who are treated with a gross tumor volume (GTV) to planning tumor volume (PTV) expansion of ≤1 cm will have progression-free survival (PFS) and overall survival (OS) rates similar to those treated in accordance with standard protocols by the Radiation Therapy Oncology Group or European Organisation for Research and Treatment of Cancer. Furthermore, the PFS and OS of subgroups within the study population will have equivalent survival outcomes with GTV1-to-PTV1 margins of 1.0 cm and 0.4 cm. METHODS AND MATERIALS Treatment plans and outcomes for patients with pathologically confirmed HGG were analyzed (n = 267). Survival (PFS and OS) was calculated from the time of the first radiation treatment and a χ2 test or Fisher exact test was used to calculate the associations between margin size and patient characteristics. Survival was estimated using Kaplan-Meier and compared using the log-rank test. All analyses were performed on the univariate level. RESULTS The median PFS and OS times were 10.6 and 19.1 months, respectively. By disease, the median PFS and OS times were 8.6 and 16.1 months for glioblastoma and 26.7 and 52.5 months for anaplastic glioma. The median follow-up time was 18.3 months. The treatment margin had no effect on outcome and the 1.0 cm GTV1-PTV1 margin subgroup (n = 212) showed median PFS and OS times of 10.7 and 19.1 months, respectively, and the 0.4 cm margin subgroup (n = 55) 10.2 and 19.3 months, respectively. In comparison with the standard treatment with 2 cm to 3 cm margins, there was not a significant difference in outcomes. CONCLUSIONS There is no apparent difference in survival when utilizing smaller versus larger margins as defined by the guidelines of the Radiation Therapy Oncology Group and European Organisation for Research and Treatment of Cancer. Although there remains no class I evidence that outcomes after treatment with smaller margins are identical to those after treatment with larger margins, this large series with long-term follow up suggests that a reduction of the margins is safe and further investigation is warranted.
Collapse
|
119
|
Le TNT, Lim H, Hamilton AM, Parkins KM, Chen Y, Scholl TJ, Ronald JA. Characterization of an Orthotopic Rat Model of Glioblastoma Using Multiparametric Magnetic Resonance Imaging and Bioluminescence Imaging. ACTA ACUST UNITED AC 2018; 4:55-65. [PMID: 30206545 PMCID: PMC6127346 DOI: 10.18383/j.tom.2018.00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor, with most patients dying within 15–18 months of diagnosis despite aggressive therapy. Preclinical GBM models are valuable for exploring GBM progression and for evaluating new therapeutics or imaging approaches. The rat C6 glioma model shares similarities with human GBM, and application of noninvasive imaging enables better study of disease progression. Here, multiparametric magnetic resonance imaging (mpMRI) and bioluminescence imaging (BLI) were applied to characterize longitudinal development of orthotopic luciferase-expressing C6 tumors. Across all rats (n = 11), a large variability was seen for BLI signal, a relative measure of C6 cell viability, but in most individuals, BLI signal peaked at day 11 and decreased thereafter. T2 and contrast-enhanced T1 tumor volumes significantly increased over time (P < .05), and volume measures did not correlate with BLI signal. After day 11, tumor regions of noncontrast enhancement appeared in postcontrast T1-weighted magnetic resonance imaging, and had significantly higher apparent diffusion coefficient values compared with contrast-enhanced regions (P < .05). This suggests formation of ill-perfused, necrotic regions beyond day 11, which were apparent at end-point–matched tissue sections. Our study represents the first combined use of BLI and mpMRI to characterize the progression of disease in the orthotopic C6 rat model, and it highlights the variability in tumor growth, the complementary information from BLI and mpMRI, and the value of multimodality imaging to better characterize tumor development. Future application of these imaging tools will be useful for evaluation of treatment response, and should be pertinent for other preclinical models.
Collapse
Affiliation(s)
- Trung N T Le
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | - Heeseung Lim
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | | | - Katie M Parkins
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | - Yuanxin Chen
- Robarts Research Institute, Western University, London, ON, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada.,Ontario Institute for Cancer Research, Toronto, ON, Canada; and
| | - John A Ronald
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
120
|
Ulutin C, Fayda M, Aksu G, Cetinayak O, Kuzhan O, Ors F, Beyzadeoglu M. Primary Glioblastoma Multiforme in Younger Patients: A Single-institution Experience. TUMORI JOURNAL 2018; 92:407-11. [PMID: 17168433 DOI: 10.1177/030089160609200507] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and Background To report our experience of patients with primary glioblastoma multiforme of young age by evaluating the characteristics, prognostic factors, and treatment outcomes. Patients and Methods Seventy patients with primary glioblastoma multiforme (GBM) treated at our department between 1996 and 2004 were studied. The male-female ratio was 2.6:1. The median age was 53 (16-74). Sixty-eight patients (97%) were operated on before radiotherapy and 2 patients (3%) underwent only stereotactic biopsy. All patients received radiotherapy. Postoperative chemotherapy as an adjuvant to radiotherapy was given to 9 patients (12%). The patients were divided into 2 groups according to their age (group A ≤35 years, n = 21 vs group B >35 years, n = 49). Survival was determined with the Kaplan-Meier method and differences were compared using the log-rank test. Cox regression analysis was performed to identify the independent prognostic factors. Karnofsky performance status (≥70 vs <70), age (≤35 vs >35 years), gender, tumor size (≤4 vs >4 cm), number of involved brain lobes (1 vs more than 1), type of surgery (total vs subtotal), preoperative seizure history (present vs absent), radiotherapy field (total cranium vs partial), total radiotherapy dose (60 vs 66 Gy), and adjuvant chemotherapy (present vs absent) were evaluated in univariate analysis. Results The median survival was 10.3 months in the whole group, 19.5 months in the younger age group and 5.7 months in the older age group. During follow-up re-craniotomy was performed in 2 patients (3%), and 1 patient (1%) developed spinal seeding metastases and was given spinal radiotherapy. In univariate analysis younger age vs older age: median 19.5 months vs 5.27 months (P = 0.0012); Karnofsky performance status ≥70 vs <70: median 15.3 months vs 2.67 months (P <0.0001), and external radiotherapy dose 60 Gy vs 66 Gy: median 11.6 months vs 3 months (P = 0.02) were found as significant prognostic factors for survival. In regression analysis a worse performance status (KPS <70) was found to be the only independent factor for survival (P = 0.014, 95% CI HR = 0.0043 [0.0001-0.15]). Conclusions Younger patients with primary glioblastoma multiforme had a relatively long survival (median, 19.5 months, with a 2-year survival rate of 30%) compared to older patients. This was due particularly to their better performance status.
Collapse
Affiliation(s)
- Cüneyt Ulutin
- Department of Radiation Oncology, GATA Hospital, Ankara, Turkey.
| | | | | | | | | | | | | |
Collapse
|
121
|
Mañé JM, Fernández R, Muñoz A, Rubio I, Ferreiro J, López-Argumedo G, Barceló R, López-Vivanco G. Preradiation Chemotherapy with VM-26 and CCNU in Patients with Glioblastoma Multiforme. TUMORI JOURNAL 2018; 90:562-6. [PMID: 15762357 DOI: 10.1177/030089160409000605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Aims and Background The objective of the study was to evaluate the efficacy of combined chemoradiation in patients with newly diagnosed glioblastoma multiforme. The main end points were time to progression and overall survival. Methods Thirty-one patients with glioblastoma multiforme underwent surgery whenever possible and then received intravenous VM26 (120 mg/m2) and oral CCNU (120 mg/m2) for three cycles followed by radiotherapy (60 Gy). Results Surgery consisted of a complete resection in 39% of patients, partial resection in 35% and a biopsy in 26%. Sixteen patients had clinical or radiological evidence of progression during or after chemotherapy. Hematologic toxicity was mild. Forty-five percent of patients received the scheduled dose of radiation. The outcome was disappointing, with a median time to progression of 18 weeks and median survival of 37.17 weeks. Conclusions The survival of patients with glioblastoma multiforme remains disappointing. Multimodal therapy does not seem to modify the evolution of the tumor. Stratification according to prognostic factors might detect a potential benefit of other therapeutic approaches.
Collapse
Affiliation(s)
- Joan M Mañé
- Medical Oncology, Hospital de Cruces, Osakidetza/Servicio Vasco de Salud, Barakaldo (Bizkaia), Spain.
| | | | | | | | | | | | | | | |
Collapse
|
122
|
Carella AM, Giordano D, Santini G, Frassoni F, Podestà M, Van Lint MT, Bacigalupo A, Nati S, Vimercati R, Occhini D, Bistolfi F, Lucarelli G, Lercari G, Marmont AM. High Dose BCNU Followed by Autologous Bone Marrow Infusion in Glioblastoma Multiforme. TUMORI JOURNAL 2018; 67:473-5. [PMID: 6275588 DOI: 10.1177/030089168106700513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Four patients with glioblastoma multiforme were treated with high dose BCNU, 800 mg/m2, and autologous marrow transplantation. Two patients were treated in their first and 2 in their second remission. All patients are alive between 1 and 12 months from transplantation.
Collapse
|
123
|
|
124
|
Yang F, Yang P, Zhang C, Wang Y, Zhang W, Hu H, Wang Z, Qiu X, Jiang T. Stratification according to recursive partitioning analysis predicts outcome in newly diagnosed glioblastomas. Oncotarget 2018; 8:42974-42982. [PMID: 28496000 PMCID: PMC5522120 DOI: 10.18632/oncotarget.17322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 12/16/2016] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma accounts for more than half of diffuse gliomas. The prognosis of patients with glioblastoma remains poor despite comprehensive and intensive treatments. Furthermore, the clinical significance of molecular parameters and routinely available clinical variables for the prognosis prediction of glioblastomas remains limited. The authors describe a novel model may help in prognosis prediction and clinical management of glioblastoma patients. We performed a recursive partitioning analysis to generate three independent prognostic classes of 103 glioblastomas patients from TCGA dataset. Class I (MGMT promoter methylated, age <58), class II (MGMT promoter methylation, age ≥58; MGMT promoter unmethylation, age <54, KPS ≥70; MGMT promoter unmethylation, age >59, KPS ≥70), class III (MGMT promoter unmethylation, age 54-58, KPS ≥70; MGMT promoter unmethylation, KPS <70). Age, KPS and MGMT promoter methylation were the most significant prognostic factors for overall survival. The results were validated in CGGA dataset. This was the first study to combine various molecular parameters and clinical factors into recursive partitioning analysis to predict the prognosis of patients with glioblastomas. We included MGMT promoter methylation in our study, which could give better suggestion to patients for their chemotherapy. This clinical study will serve as the backbone for the future incorporation of molecular prognostic markers currently in development. Thus, our recursive partitioning analysis model for glioblastomas may aid in clinical prognosis evaluation.
Collapse
Affiliation(s)
- Fan Yang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Pei Yang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chuanbao Zhang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yongzhi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huimin Hu
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhiliang Wang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaoguang Qiu
- Department of Radiation Therapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
125
|
Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, Siemianowicz K, Likus W, Wiechec E, Toyota BD, Hoshyar R, Seyfoori A, Sepehri Z, Ande SR, Khadem F, Akbari M, Gorman AM, Samali A, Klonisch T, Ghavami S. Glioblastoma and chemoresistance to alkylating agents: Involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther 2018; 184:13-41. [DOI: 10.1016/j.pharmthera.2017.10.017] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
126
|
Ozdemir-Kaynak E, Qutub AA, Yesil-Celiktas O. Advances in Glioblastoma Multiforme Treatment: New Models for Nanoparticle Therapy. Front Physiol 2018; 9:170. [PMID: 29615917 PMCID: PMC5868458 DOI: 10.3389/fphys.2018.00170] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 02/20/2018] [Indexed: 11/30/2022] Open
Abstract
The most lethal form of brain cancer, glioblastoma multiforme, is characterized by rapid growth and invasion facilitated by cell migration and degradation of the extracellular matrix. Despite technological advances in surgery and radio-chemotherapy, glioblastoma remains largely resistant to treatment. New approaches to study glioblastoma and to design optimized therapies are greatly needed. One such approach harnesses computational modeling to support the design and delivery of glioblastoma treatment. In this paper, we critically summarize current glioblastoma therapy, with a focus on emerging nanomedicine and therapies that capitalize on cell-specific signaling in glioblastoma. We follow this summary by discussing computational modeling approaches focused on optimizing these emerging nanotherapeutics for brain cancer. We conclude by illustrating how mathematical analysis can be used to compare the delivery of a high potential anticancer molecule, delphinidin, in both free and nanoparticle loaded forms across the blood-brain barrier for glioblastoma.
Collapse
Affiliation(s)
- Elif Ozdemir-Kaynak
- Department of Bioengineering, Faculty of Engineering, Ege University, Bornova-Izmir, Turkey
| | - Amina A Qutub
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, Bornova-Izmir, Turkey.,Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, United States
| |
Collapse
|
127
|
Ali AN, Zhang P, Yung WKA, Chen Y, Movsas B, Urtasun RC, Jones CU, Choi KN, Michalski JM, Fischbach AJ, Markoe AM, Schultz CJ, Penas-Prado M, Garg MK, Hartford AC, Kim HE, Won M, Curran WJ. NRG oncology RTOG 9006: a phase III randomized trial of hyperfractionated radiotherapy (RT) and BCNU versus standard RT and BCNU for malignant glioma patients. J Neurooncol 2018; 137:39-47. [PMID: 29404979 DOI: 10.1007/s11060-017-2558-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/27/2017] [Indexed: 11/28/2022]
Abstract
From 1990 to 1994, patients with newly diagnosed malignant gliomas were enrolled and randomized between hyperfractionated radiation (HFX) of 72.0 Gy in 60 fractions given twice daily and 60.0 Gy in 30 fractions given once daily. All patients received 80 mg/m2 of 1,3 bis(2 chloroethyl)-1 nitrosourea on days 1-3 q8 weeks for 1 year. Patients were stratified by age, KPS, and histology. The primary endpoint was overall survival (OS), with secondary endpoints including progression-free survival (PFS) and toxicity. Out of the 712 patients accrued, 694 (97.5%) were analyzable cases (350 HFX, 344 standard arm). There was no significant difference between the arms on overall acute or late treatment-related toxicity. No statistically significant effect for HFX, as compared to standard therapy, was found on either OS, with a median survival time (MST) of 11.3 versus 13.1 months (p = 0.20) or PFS, with a median PFS time of 5.7 versus 6.9 months (p = 0.18). The treatment effect on OS remained insignificant based on the multivariate analysis (hazard ratio 1.16; p = 0.0682). When OS was analyzed by histology subgroup there was also no significant difference between the two arms for patients with glioblastoma multiforme (MST: 10.3 vs. 11.2 months; p = 0.34), anaplastic astrocytoma (MST: 69.8 vs. 50.0 months; p = 0.91) or anaplastic oligodendroglioma (MST: 92.1 vs. 66.5 months; p = 0.33). Though this trial provided many invaluable secondary analyses, there was no trend or indication of a benefit to HFX radiation to 72.0 Gy in any subset of malignant glioma patients.
Collapse
Affiliation(s)
- Arif N Ali
- Emory University/Winship Cancer Institute, 1365 Clifton Rd NE, Atlanta, GA, 30322, USA.
| | - Peixin Zhang
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA, USA
| | - W K Alfred Yung
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuhchyau Chen
- University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin Movsas
- Henry Ford Hospital accruals Fox Chase Cancer Center, Detroit, MI, USA
| | | | - Christopher U Jones
- Sutter General Hospital accruals Radiological Associates of Sacramento, Sacramento, CA, USA
| | - Kwang N Choi
- State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | | | | | - Arnold M Markoe
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | | | - Madhur K Garg
- Montefiore Medical Center, Moses Campus, Bronx, NY, USA
| | | | | | - Minhee Won
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA, USA
| | - Walter J Curran
- Emory University/Winship Cancer Institute, 1365 Clifton Rd NE, Atlanta, GA, 30322, USA
| |
Collapse
|
128
|
Kamran N, Alghamri MS, Nunez FJ, Shah D, Asad AS, Candolfi M, Altshuler D, Lowenstein PR, Castro MG. Current state and future prospects of immunotherapy for glioma. Immunotherapy 2018; 10:317-339. [PMID: 29421984 PMCID: PMC5810852 DOI: 10.2217/imt-2017-0122] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/30/2017] [Indexed: 12/14/2022] Open
Abstract
There is a large unmet need for effective therapeutic approaches for glioma, the most malignant brain tumor. Clinical and preclinical studies have enormously expanded our knowledge about the molecular aspects of this deadly disease and its interaction with the host immune system. In this review we highlight the wide array of immunotherapeutic interventions that are currently being tested in glioma patients. Given the molecular heterogeneity, tumor immunoediting and the profound immunosuppression that characterize glioma, it has become clear that combinatorial approaches targeting multiple pathways tailored to the genetic signature of the tumor will be required in order to achieve optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Kamran
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Felipe J Nunez
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Diana Shah
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Antonela S Asad
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - David Altshuler
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Maria G Castro
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| |
Collapse
|
129
|
Yaman E, Buyukberber S, Uner A, Coskun U, Yamac D, Ozturk B, Kaya AO, Yildiz R. Carboplatin and oral cyclophosphamide combination after temozolomide failure in malignant gliomas. TUMORI JOURNAL 2018; 94:674-80. [DOI: 10.1177/030089160809400505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Temozolomide is a novel cytotoxic agent for malignant gliomas. However, treatment failure occurs approximately in half of patients, and the optimal regimen in this setting has yet to be defined. In the present study, we assessed retrospectively the efficacy and toxicity of the combination of carboplatin and oral cyclophosphamide in temozolomide-resistant patients. Methods We evaluated the medical records of 30 patients with malignant gliomas. After failure of temozolomide therapy, patients were treated with a combination of carboplatin and oral cyclophosphamide. Treatment consisted of intravenous carboplatin AUC 6 (based on the Calvert Formula) on day 1 and oral cyclophosphamide 75 mg/m2 daily on days 1 to 14, followed by 14 days of rest, with the treatment repeated every 4 weeks. Results All patients were evaluated for response and toxicity. The objective response rate was 30%, including 9 partial responses. Median time to disease progression and median overall survival was 7 months and 8 months, respectively. Clinically responsive patients had statistically significant longer progression-free survival and overall survival than unresponsive patients. Hematological side effects were commonly observed toxicities, with neutropenia the most frequent. Conclusions Our data suggest that carboplatin and oral cyclophosphamide therapy is a convenient regimen after failure of temozolomide therapy in patients with malignant gliomas because of its activity, feasibility and tolerability. Further prospective studies are needed in this setting.
Collapse
Affiliation(s)
| | - Emel Yaman
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Suleyman Buyukberber
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Aytug Uner
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ugur Coskun
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Deniz Yamac
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Banu Ozturk
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ali Osman Kaya
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ramazan Yildiz
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
130
|
Nagy M, Schulz-Ertner D, Bischof M, Welzel T, Hof H, Debus J, Combs SE. Long-Term Outcome of Postoperative Irradiation in Patients with Newly Diagnosed WHO Grade III Anaplastic Gliomas. TUMORI JOURNAL 2018; 95:317-24. [DOI: 10.1177/030089160909500308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose Patients with anaplastic gliomas have a more favorable overall survival than patients with glioblastomas. In most analyses, WHO grade III and IV tumors are not analyzed separately. The present analysis reports outcome after postoperative radiotherapy in patients with WHO grade III gliomas. Patients and methods Between January 1988 and January 2007, 127 patients with WHO grade III tumors were treated with radiotherapy; the histological classification was pure astrocytoma in 104 patients, oligoastrocytoma in 12 and pure oligodendroglioma in 11 patients. Median age was 48 years. After the primary diagnosis, a biopsy had been performed in 72 patients; subtotal and total resections were performed in 37 and 18 patients, respectively. In all patients radiotherapy was applied with a median dose of 60 Gy in conventional fractionation. The median follow-up time was 18 months. Results Median overall survival was 17 months. Overall survival was significantly influenced by the extent of surgery. Median overall survival was 32 months after complete resection, 36 months after subtotal resection, and 12 months after biopsy. Median overall survival was 7 months for patients with anaplastic astrocytomas, 44 months for patients with mixed tumors, and 47 months for those with pure oligodendrogliomas. Age significantly influenced overall survival. Median progression-free survival was 9 months; the extent of neurosurgical resection significantly influenced progression-free survival. Conclusion Patients with WHO grade III anaplastic astrocytomas, oligodendrogliomas and oligoastrocytomas show favorable overall survival after postoperative radiotherapy compared with glioblastoma patients and should therefore be analyzed separately. Radiochemotherapy might further improve outcome.
Collapse
Affiliation(s)
- Monika Nagy
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Daniela Schulz-Ertner
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Marc Bischof
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Thomas Welzel
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Holger Hof
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
131
|
Donato V, Papaleo A, Castrichino A, Banelli E, Giangaspero F, Salvati M, Delfini R. Prognostic Implication of Clinical and Pathologic Features in Patients with Glioblastoma Multiforme Treated with Concomitant Radiation plus Temozolomide. TUMORI JOURNAL 2018; 93:248-56. [PMID: 17679459 DOI: 10.1177/030089160709300304] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Glioblastoma multiforme is the most common and most malignant primary brain tumor in adults. The current standard of care for glioblastoma is surgical resection to the extent feasible, followed by adjuvant radiotherapy plus temozolomide, given concomitantly with and after radiotherapy. This report is a prospective observational study of 43 cases treated in the Department of Radiotherapy, University of Rome La Sapienza, Italy. We examine the relationship between pathologic features and objective response rate in adult patients treated with concomitant radiation plus temozolomide to identify clinical, neuroradiologic, pathologic, and molecular factors with prognostic significance. Methods Forty-three consecutive patients (24 males and 19 females), ages 15-77 years (median, 57) with newly diagnosed glioblastoma multiforme, were included in this trial between 2002 and 2004 at our department. All patients were treated with surgery (complete resection in 81%, incomplete in 19%) followed by concurrent temozolomide (75 mg/m2/day) and radiotherapy (median tumor dose, 60 Gy), followed by temozolomide, 200 mg/m2/day for 5 consecutive days every 28 days. Neurologic evaluations were performed monthly and cranial magnetic resonance bimonthly. We analyzed age, clinical manifestations at diagnosis, seizures, Karnofsky performance score, tumor location, extent of resection, proliferation index (Ki-67 expression), p53, platelet-derived growth factor and epidermal growth factor receptor immunohistochemical expression as prognostic factors in the patients. The Kaplan-Meier statistical method and logrank test were used to assess correlation with survival. Results Fourteen patients (32%) manifested clinical and neuroradiographic evidence of tumor progression within 6 months of surgery. In contrast, 5 patients (12%) showed no disease progression for 18 months from the beginning of treatment. Median overall survival was 19 months. Multivariate analysis revealed that an age of 60 years or older (P <0.03), a postoperative performance score ≤70 (P = 0.04), the nontotal tumor resection (P = 0.03), tumor size >4 cm (P = 0.01) and proliferation index overexpression (P = 0.001) were associated with the worst prognosis. p53, PDGF and EGFR overexpression were not significant prognostic factors associated with survival. Conclusions The results suggest that analysis of prognostic markers in glioblastoma multiforme is complex. In addition to previously recognized prognostic variables such as age and Karnofsky performance score, tumor size, total resection and proliferation index overexpression were identified as predictors of survival in a series of patients with glioblastoma multiforme.
Collapse
Affiliation(s)
- Vittorio Donato
- Department of Radiotherapy, University of Rome La Sapienza, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
132
|
Mann J, Ramakrishna R, Magge R, Wernicke AG. Advances in Radiotherapy for Glioblastoma. Front Neurol 2018; 8:748. [PMID: 29379468 PMCID: PMC5775505 DOI: 10.3389/fneur.2017.00748] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/27/2017] [Indexed: 11/13/2022] Open
Abstract
External beam radiotherapy (RT) has long played a crucial role in the treatment of glioblastoma. Over the past several decades, significant advances in RT treatment and image-guidance technology have led to enormous improvements in the ability to optimize definitive and salvage treatments. This review highlights several of the latest developments and controversies related to RT, including the treatment of elderly patients, who continue to be a fragile and vulnerable population; potential salvage options for recurrent disease including reirradiation with chemotherapy; the latest imaging techniques allowing for more accurate and precise delineation of treatment volumes to maximize the therapeutic ratio of conformal RT; the ongoing preclinical and clinical data regarding the combination of immunotherapy with RT; and the increasing evidence of cancer stem-cell niches in the subventricular zone which may provide a potential target for local therapies. Finally, continued development on many fronts have allowed for modestly improved outcomes while at the same time limiting toxicity.
Collapse
Affiliation(s)
- Justin Mann
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Rohan Ramakrishna
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - A Gabriella Wernicke
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
133
|
Tini P, Nardone V, Pastina P, Battaglia G, Miracco C, Carbone SF, Sebaste L, Rubino G, Cerase A, Pirtoli L. Epidermal Growth Factor Receptor Expression Predicts Time and Patterns of Recurrence in Patients with Glioblastoma After Radiotherapy and Temozolomide. World Neurosurg 2018; 109:e662-e668. [DOI: 10.1016/j.wneu.2017.10.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 10/18/2022]
|
134
|
Zhao J, Zhu J, Lv X, Xing J, Liu S, Chen C, Xu Y. Curcumin potentiates the potent antitumor activity of ACNU against glioblastoma by suppressing the PI3K/AKT and NF-κB/COX-2 signaling pathways. Onco Targets Ther 2017; 10:5471-5482. [PMID: 29180881 PMCID: PMC5695266 DOI: 10.2147/ott.s149708] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Glioblastoma (GBM) is a highly invasive and challenging primary tumor of the central nervous system (CNS), and currently available treatments provide limited benefits to patients with this disease. Therefore, the development of novel therapeutic targets and effective treatment strategies is essential. Nimustine hydrochloride (ACNU) is widely used as the standard chemotherapeutic agent and is frequently administered together with other chemotherapeutic agents in clinical studies. Curcumin, a natural polyphenolic compound, could potentially be combined with chemotherapeutics for cancer treatment; however, there are no reports of studies where ACNU and curcumin were combined for GBM treatment, and the mechanisms underlying their activity remain poorly understood. In the present study, we investigated the effects of combined treatment with curcumin and ACNU on GBM cells and found that it significantly enhanced the inhibition of cell proliferation, colony formation, migration, and invasion. In addition, co-treatment with curcumin increased ACNU-induced apoptosis through enhancing the release of cytochrome c from the mitochondrial intermembrane space into the cytosol. Further, curcumin and ACNU acted synergistically in their antitumor effects by targeting N-cadherin/MMP2/9, PI3K/AKT, and NF-κB/COX-2 signaling. These results indicate that curcumin can enhance the anti-proliferation, anti-migration, and proapoptotic activities of ACNU against GBM, and provide strong evidence that combined treatment with curcumin and ACNU has the potential to be an effective therapeutic option for GBM.
Collapse
Affiliation(s)
| | - Jiabin Zhu
- Department of Urology, First Affiliated Hospital of Dalian Medical University
| | | | | | - Shuang Liu
- Department of Gastroenterology, Second Affiliated Hospital of Dalian Medical University
| | - Chen Chen
- Department of Cardiovascular, First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | | |
Collapse
|
135
|
Lam S, Lin Y, Zinn P, Su J, Pan IW. Patient and treatment factors associated with survival among pediatric glioblastoma patients: A Surveillance, Epidemiology, and End Results study. J Clin Neurosci 2017; 47:285-293. [PMID: 29102237 DOI: 10.1016/j.jocn.2017.10.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/10/2017] [Indexed: 10/18/2022]
Abstract
Glioblastoma (GBM) is a rare malignancy in children. The United States Surveillance, Epidemiology, and End Results (SEER) database allows large-scale analyses of clinical characteristics and prognostic features. We used it to study patients aged <20 years with histologically confirmed GBM (2000-2010) and examined the relationship between patient demographics, tumor characteristics, patterns of treatment, and outcomes. The primary outcome was disease-specific survival. 302 subjects were identified, with median age 11 years. Median follow-up was 32 months (95% CI 27-39). 34.4% had gross total resection (GTR). 61% underwent radiation after surgery (17% of subjects <3 years, 67% of those aged 4-19 years). Median survival and 2-year survival rates were 20 months and 46.9%, respectively. In multivariate analyses, age, tumor location, extent of resection, and year of diagnosis were significantly associated with the primary outcome. Compared to those aged 0-4 years, subjects aged 5-9 years and 10-14 years had higher risk of mortality. Infratentorial tumor location (HR 2.0, 95% CI 1.2-3.3, p = 0.007) and subtotal resection (HR 2.04, 95% CI 1.4-3.0, p < 0.001) were associated with increased mortality. Later year of diagnosis was significantly associated with decreased risk of death (HR 0.93, 95% CI 0.9-0.99, p = 0.031). There was no association between sex, race, region, or tumor size and the primary outcome. Repeat analyses examining all-cause mortality identified the same risk factors as for CNS cancer-specific mortality. Younger age, supratentorial location, GTR, and later year of diagnosis were associated with improved survival.
Collapse
Affiliation(s)
- Sandi Lam
- Baylor College of Medicine, Department of Neurosurgery, Houston, TX, USA; Texas Children's Hospital, Division of Pediatric Neurosurgery, Houston, TX, USA.
| | - Yimo Lin
- Baylor College of Medicine, Department of Neurosurgery, Houston, TX, USA; Texas Children's Hospital, Division of Pediatric Neurosurgery, Houston, TX, USA
| | - Pascal Zinn
- Baylor College of Medicine, Department of Neurosurgery, Houston, TX, USA; Texas Children's Hospital, Division of Pediatric Neurosurgery, Houston, TX, USA
| | - Jack Su
- Baylor College of Medicine, Department of Pediatrics, Houston, TX, USA; Texas Children's Hospital, Division of Pediatric Hematology/Oncology, Houston, TX, USA
| | - I-Wen Pan
- Baylor College of Medicine, Department of Neurosurgery, Houston, TX, USA; Texas Children's Hospital, Division of Pediatric Neurosurgery, Houston, TX, USA
| |
Collapse
|
136
|
Balañá C, Alonso M, Hernandez-Lain A, Hernandez A, Perez-Segura P, Pineda E, Ramos A, Sanchez AR, Teixidor P, Verger E, Benavides M. SEOM clinical guidelines for anaplastic gliomas (2017). Clin Transl Oncol 2017; 20:16-21. [PMID: 29058264 PMCID: PMC5785606 DOI: 10.1007/s12094-017-1762-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 11/29/2022]
Abstract
The SEOM/GEINO clinical guidelines provide recommendations for radiological, and molecular diagnosis, treatment and follow-up of adult patients with anaplastic gliomas (AG). We followed the 2016 WHO classification which specifies the major diagnostic/prognostic and predictive value of IDH1/IDH2 missense mutations and 1p/19q codeletions in AG. The diagnosis of anaplastic oligoastrocytoma is discouraged. Surgery, radiotherapy and chemotherapy with PCV or TMZ are the first-line standard of care for AG with slight modifications according to molecular variables. A multidisciplinary team is highly recommended in the management of these tumors.
Collapse
Affiliation(s)
- C Balañá
- Institut Català Oncologia Badalona, Ct. Canyet, s/n, 08916, Barcelona, Spain.
| | - M Alonso
- Complejo Hospitalario Virgen del Rocío, Seville, Spain
| | | | | | - P Perez-Segura
- Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - E Pineda
- Hospital Clínic i Provincial, Barcelona, Spain
| | - A Ramos
- Hospital 12 de Octubre, Madrid, Spain
| | - A R Sanchez
- Complejo Asistencial Universitario de León, León, Spain
| | - P Teixidor
- Hospital Universitari Germans Trias i Pujol Badalona, Barcelona, Spain
| | - E Verger
- Hospital Clínic i Provincial, Barcelona, Spain
| | - M Benavides
- Hospital Universitario Regional y Virgen de la Victoria, Málaga, Spain
| |
Collapse
|
137
|
Nagy Á, Garzuly F, Padányi G, Szűcs I, Feldmann Á, Murnyák B, Hortobágyi T, Kálmán B. Molecular Subgroups of Glioblastoma- an Assessment by Immunohistochemical Markers. Pathol Oncol Res 2017; 25:21-31. [PMID: 28948518 DOI: 10.1007/s12253-017-0311-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/15/2017] [Indexed: 12/11/2022]
Abstract
Comprehensive molecular characterization of and novel therapeutic approaches to glioblastoma have been explored as a result of advancements in biotechnologies. In this study, we aimed to bring basic research discoveries closer to clinical practice and ultimately incorporate molecular classification into the routine histopathological evaluation of grade IV gliomas. Integrated results of genome-wide sequencing, transcriptomic and epigenomic analyses by The Cancer Genome Atlas Network defined the classic, proneural, neural and mesenchymal subtypes of this tumor. In a retrospective cohort, we analyzed selected subgroup-defining molecular markers in formalin-fixed paraffin-embedded surgical specimens by immunohistochemistry. Quantitative and qualitative scores of marker expression were tested in hierarchical cluster analyses to evaluate segregations of the molecular subgroups, which then were correlated with clinical parameters including patients' age, gender and overall survival. Our study has confirmed the separation of molecular glioblastoma subgroups with clear trends regarding clinical correlations. Future analyses in a larger, prospective cohort using similar methods are expected to facilitate the development of a molecular diagnostic panel that may complement routine histological work up and support prognostication as well as treatment decisions in glioblastoma.
Collapse
Affiliation(s)
- Ádám Nagy
- Faculty of Health Sciences, School of Graduate Studies, University of Pécs, Pécs, Hungary
| | - Ferenc Garzuly
- Markusovszky University Teaching Hospital, University of Pecs, 5. Markusovszky Street, Szombathely, 9700, Hungary
| | - Gergely Padányi
- Markusovszky University Teaching Hospital, University of Pecs, 5. Markusovszky Street, Szombathely, 9700, Hungary
| | | | - Ádám Feldmann
- Faculty of Medicine, Institute of Behavioral Sciences, University of Pécs, Pécs, Hungary
| | - Balázs Murnyák
- Department of Pathology, Division of Neuropathology, University of Debrecen, Debrecen, Hungary
| | - Tibor Hortobágyi
- Department of Pathology, Division of Neuropathology, University of Debrecen, Debrecen, Hungary
| | - Bernadette Kálmán
- Faculty of Health Sciences, School of Graduate Studies, University of Pécs, Pécs, Hungary. .,Markusovszky University Teaching Hospital, University of Pecs, 5. Markusovszky Street, Szombathely, 9700, Hungary.
| |
Collapse
|
138
|
Hohmann T, Grabiec U, Vogel C, Ghadban C, Ensminger S, Bache M, Vordermark D, Dehghani F. The Impact of Non-Lethal Single-Dose Radiation on Tumor Invasion and Cytoskeletal Properties. Int J Mol Sci 2017; 18:E2001. [PMID: 28926987 PMCID: PMC5618650 DOI: 10.3390/ijms18092001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 12/27/2022] Open
Abstract
Irradiation is the standard therapy for glioblastoma multiforme. Glioblastoma are highly resistant to radiotherapy and the underlying mechanisms remain unclear. To better understand the biological effects of irradiation on glioblastoma cells, we tested whether nonlethal irradiation influences the invasiveness, cell stiffness, and actin cytoskeleton properties. Two different glioblastoma cell lines were irradiated with 2 Gy and changes in mechanical and migratory properties and alterations in the actin structure were measured. The invasiveness of cell lines was determined using a co-culture model with organotypic hippocampal slice cultures. Irradiation led to changes in motility and a less invasive phenotype in both investigated cell lines that were associated with an increase in a "generalized stiffness" and changes in the actin structure. In this study we demonstrate that irradiation can induce changes in the actin cytoskeleton and motility, which probably results in reduced invasiveness of glioblastoma cell lines. Furthermore, "generalized stiffness" was shown to be a profound marker of the invasiveness of a tumor cell population in our model.
Collapse
Affiliation(s)
- Tim Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Urszula Grabiec
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Carolin Vogel
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Chalid Ghadban
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Stephan Ensminger
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120 Halle, Germany.
| | - Matthias Bache
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120 Halle, Germany.
| | - Dirk Vordermark
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120 Halle, Germany.
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| |
Collapse
|
139
|
Hu H, Wang Z, Li M, Zeng F, Wang K, Huang R, Wang H, Yang F, Liang T, Huang H, Jiang T. Gene Expression and Methylation Analyses Suggest DCTD as a Prognostic Factor in Malignant Glioma. Sci Rep 2017; 7:11568. [PMID: 28912488 PMCID: PMC5599690 DOI: 10.1038/s41598-017-11962-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/31/2017] [Indexed: 11/09/2022] Open
Abstract
Malignant glioma is the most common brain cancer with dismal outcomes. Individual variation of the patients’ survival times is remarkable. Here, we investigated the transcriptome and promoter methylation differences between patients of malignant glioma with short (less than one year) and the patients with long (more than three years) survival in CGGA (Chinese Glioma Genome Atlas), and validated the differences in TCGA (The Cancer Genome Atlas) to identify the genes whose expression levels showed high concordance with prognosis of glioma patients, as well as played an important role in malignant progression. The gene coding a key enzyme in genetic material synthesis, dCMP deaminase (DCTD), was found to be significantly correlated with overall survival and high level of DCTD mRNA indicated shorter survival of the patients with malignant glioma in different databases. Our finding revealed DCTD as an efficient prognostic factor for malignant glioma. As DCTD inhibitor gemcitabine has been proposed as an adjuvant therapy for malignant glioma, our finding also suggests a therapeutic value of gemcitabine for the patients with high expression level of DCTD.
Collapse
Affiliation(s)
- Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Zheng Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Mingyang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Kuanyu Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | | | - Fan Yang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Tingyu Liang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Hua Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China. .,Chinese Glioma Cooperative Group (CGCG), Beijing, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China. .,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
140
|
Chan DTM, Yi-Pin Sonia H, Poon WS. 5-Aminolevulinic acid fluorescence guided resection of malignant glioma: Hong Kong experience. Asian J Surg 2017; 41:467-472. [PMID: 28844780 DOI: 10.1016/j.asjsur.2017.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/19/2017] [Accepted: 06/28/2017] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND 5-Aminolevulinic Acid (5-ALA) induced fluorescence is useful in guiding glioma resection. The extent of 5-ALA accumulation is beyond gadolinium contrast enhancement.1,2 Supratotal resection may be achieved, potentially granting patients with better survival. We present our experience on 5-ALA guided glioma resection in Chinese ethnics. METHOD Sixteen Patients ingested 5-ALA (Gliolan, Medas Germany) 20 mg/kg·m2 4 h before surgery. The tumor resection was guided by fluorescence with neurosurgical microscope. Patient was monitored for general condition, especially for new neurological deficits. Postoperative MRI served as the assessment for extent of resection (EOR). RESULT High grade glioma was confirmed in 12 cases, low grade glioma in three and one inflammation. 5-ALA was used in ten patients with known malignant glioma, and in six patients with presumed diagnosis of malignant glioma. Fifteen cases had positive fluorescence. The intensity was strong in eight and moderate in seven cases. MRI suggested total resection was achieved in 9 patients, near total resection in two and five had subtotal resection. EOR was associated with duration between ingestion of 5-ALA and timing when microscope was brought in for visualization of fluorescence (p = 0.038). Two patients suffered from temporary visual field defects. One patient developed hemiparesis after surgery. CONCLUSION 5-ALA is a useful intra-operative guidance for resection. It increases the percentage of total removal of the tumor. It should be used within the window period of the action (4-12 h).
Collapse
Affiliation(s)
- Danny Tat Ming Chan
- CUHK Otto Wong Brain Tumor Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR.
| | - Hsieh Yi-Pin Sonia
- CUHK Otto Wong Brain Tumor Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR
| | - Wai Sang Poon
- CUHK Otto Wong Brain Tumor Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR
| |
Collapse
|
141
|
McCarthy DJ, Komotar RJ, Starke RM, Connolly ES. Randomized Trial for Short-Term Radiation Therapy With Temozolomide in Elderly Patients With Glioblastoma. Neurosurgery 2017; 81:N21-N23. [PMID: 28859461 DOI: 10.1093/neuros/nyx399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- David J McCarthy
- Department of Neurosurgery University of Miami Miller School of Medicine Miami, Florida
| | - Ricardo J Komotar
- Department of Neurosurgery University of Miami Miller School of Medicine Miami, Florida
| | - Robert M Starke
- Department of Neurosurgery University of Miami Miller School of Medicine Miami, Florida
| | - E Sander Connolly
- Department of Neurological Surgery Columbia University College of Physicians and Surgeons New York, New York
| |
Collapse
|
142
|
Sekerdag E, Lüle S, Bozdağ Pehlivan S, Öztürk N, Kara A, Kaffashi A, Vural I, Işıkay I, Yavuz B, Oguz KK, Söylemezoğlu F, Gürsoy-Özdemir Y, Mut M. A potential non-invasive glioblastoma treatment: Nose-to-brain delivery of farnesylthiosalicylic acid incorporated hybrid nanoparticles. J Control Release 2017; 261:187-198. [PMID: 28684169 DOI: 10.1016/j.jconrel.2017.06.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/29/2017] [Accepted: 06/28/2017] [Indexed: 11/26/2022]
Abstract
New drug delivery systems are highly needed in research and clinical area to effectively treat gliomas by reaching a high antineoplastic drug concentration at the target site without damaging healthy tissues. Intranasal (IN) administration, an alternative route for non-invasive drug delivery to the brain, bypasses the blood-brain-barrier (BBB) and eliminates systemic side effects. This study evaluated the antitumor efficacy of farnesylthiosalicylic acid (FTA) loaded (lipid-cationic) lipid-PEG-PLGA hybrid nanoparticles (HNPs) after IN application in rats. FTA loaded HNPs were prepared, characterized and evaluated for cytotoxicity. Rat glioma 2 (RG2) cells were implanted unilaterally into the right striatum of female Wistar rats. 10days later, glioma bearing rats received either no treatment, or 5 repeated doses of 500μM freshly prepared FTA loaded HNPs via IN or intravenous (IV) application. Pre-treatment and post-treatment tumor sizes were determined with MRI. After a treatment period of 5days, IN applied FTA loaded HNPs achieved a significant decrease of 55.7% in tumor area, equal to IV applied FTA loaded HNPs. Herewith, we showed the potential utility of IN application of FTA loaded HNPs as a non-invasive approach in glioblastoma treatment.
Collapse
Affiliation(s)
- Emine Sekerdag
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Neuroscience Research Lab, Research Center for Translational Medicine, Koҫ University, Istanbul, Turkey.
| | - Sevda Lüle
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; Neuroscience Center and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sibel Bozdağ Pehlivan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Naile Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Aslı Kara
- Department of Nanotechnology and Nanomedicine, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Department of Biology, Faculty of Art and Science, Hitit University, Çorum, Turkey
| | - Abbas Kaffashi
- Department of Nanotechnology and Nanomedicine, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Imran Vural
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ilkay Işıkay
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Burҫin Yavuz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Kader Karlı Oguz
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Figen Söylemezoğlu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Yasemin Gürsoy-Özdemir
- Neuroscience Research Lab, Research Center for Translational Medicine, Koҫ University, Istanbul, Turkey; Department of Neurology, School of Medicine, Koҫ University, Istanbul, Turkey
| | - Melike Mut
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
143
|
Jain A, Correia J, Schweder P, McMahon A, Merola J, Aspoas R. Analysis of Outcomes of Multidisciplinary Management of Gliosarcoma: A Single-Center Study, 2000-2013. World Neurosurg 2017. [PMID: 28642179 DOI: 10.1016/j.wneu.2017.06.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Gliosarcoma is a rare tumor of the central nervous system with a reported incidence of ∼2%-8% of all gliomas. We reviewed the outcomes of patients treated at our institution over a 14-year period from 2000 to 2013 to characterize overall survival (OS) and progression-free survival as well as to elucidate the additive effect of chemoradiotherapy. METHODS From January 1, 2000 to December 31, 2013, we retrospectively reviewed the clinical notes of all patients treated at our institution with a histopathologic diagnosis of gliosarcoma. This review yielded 21 patients whose clinicoradiologic data were analyzed with respect to age, sex, ethnicity, preoperative/postoperative Glasgow Coma Scale and Karnofsky Performance Scale, location, extent of resection, methylguanine DNA methyl transferase methylation status, and administration of adjuvant therapy. RESULTS The median age was 58 years (range, 40-80 years) with a male preponderance (1.6:1). Tumor location was mainly temporal (n = 6) but also parietal (n = 5), frontal (n = 4), multilobar (n = 4), and cerebellar (n = 1). Surgical resection was deemed to be total in 15 patients and subtotal in 6 patients. Methylguanine DNA methyl transferase methylation status was available for only 5 patients, with a methylation rate of 60% (3/5) and no impact on survival. Nine patients received both radiotherapy and chemotherapy (OS, 7.9 months), 7 received radiotherapy only (OS, 5.7 months), and 5 patients received no adjuvant therapy (OS, 1.4 months). The overall median survival was 5.7 months (range, 1-21.5 months) and median progression-free survival was 5 months (range, 1.4-12.4 months). CONCLUSIONS Despite an overall poor prognosis, a multimodality approach aiming for complete resection followed by radiotherapy and chemotherapy appears to be associated with better outcomes.
Collapse
Affiliation(s)
- Abhinav Jain
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Jason Correia
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Patrick Schweder
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Adele McMahon
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Joseph Merola
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Robert Aspoas
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand.
| |
Collapse
|
144
|
Lai X, Deng Z, Guo H, Zhu X, Tu W. HP1α is highly expressed in glioma cells and facilitates cell proliferation and survival. Biochem Biophys Res Commun 2017. [PMID: 28623138 DOI: 10.1016/j.bbrc.2017.06.056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Epigenetic alteration plays critical roles in gliomagenesis by regulating gene expression through modifications of Histones and DNA. Trimethylation of H3K9, an essential repressed transcription mark, and one of its methyltransferase, SUV39H1, are implicated in glioma pathogenesis and progression. We find that the protein level of HP1α, a reader of H3K9me3 is elevated in cultured glioma cell lines and glioma tissues. H3K9me3 is also upregulated. Depletion of HP1α and SUV39H1 weakens glioma cell proliferation capacity and results in apoptosis of cells. Furthermore, we find that HP1α and H3K9me3 are enriched in the FAS and PUMA promoters, which suggests that upregulated HP1α and H3K9me3 contribute to cell survival by suppressing apoptotic activators. These data suggests that up-regulated HP1α and H3K9me3 in glioma cells are functionally associated with glioma pathogenesis and progression and may serve as novel biomarkers for diagnostic and therapeutic targeting of brain tumors.
Collapse
Affiliation(s)
- Xianliang Lai
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhifeng Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wei Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
145
|
Darmon I, Morisse MC, Coutte A, Blonski M, Le Rhun E, Taillandier L, Roufai DB, Desenclos C, Trudel S, Faivre JC, Blanchard N, Chauffert B, Boone M. Temozolomide and Bevacizumab Induction before Chemoradiotherapy in Patients with Bulky Glioblastoma and/or with Severe Neurological Impairment. J Cancer 2017. [PMID: 28638456 PMCID: PMC5479247 DOI: 10.7150/jca.18339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background.New approaches are needed for patients newly diagnosed with bulky glioblastoma (GB) and/or with severe neurological impairment that cannot benefit from first line temozolomide (TMZ)-based chemoradiotherapy. Bevacizumab (BEV), an antiangiogenic anti-VEGF-R monoclonal antibody, has a rapid impact on tumor-related brain edema in recurrent GB. The present study reports the feasibility and efficacy of an induction treatment with TMZ and BEV to alleviate the initial neurological impairment and/or to reduce the tumor volume before a delayed chemoradiotherapy. Methods.We retrospectively analyzed tumor and target volumes and clinical neurological status in 39 patients with bulky GB and/or with severe neurological impairment after an induction treatment combining TMZ and BEV. Neurological and radiological responses were assessed according to RANO criteria. Calculating gross tumor and clinical target volumes (GTV and CTV) was done at diagnosis and before radiotherapy. Progression-free survival (PFS) and overall survival (OS) were determined by Kaplan Meier methods. Safety was reported according to NCTCAE. Results. A cohort of 39 patients was analyzed between December 2010 and April 2014. Upfront standard TMZ-based chemoradiotherapy was recused due either to tumor volume or impairment of neurological status and/or performance status. After TMZ/BEV induction (median time of 3 months), 6 (15%) patients achieved a partial response (PR), and 17 (44%) had a stable disease. 24 patients (62%) received a radical-intent chemoradiotherapy. TMZ-BEV induced median reduction of the clinical target volume (CTV) was 25.9% [-84.4%; - 4.8%]. The median PFS and OS were 8.4 months [95% CI: (6.6 - 9.9)] and 11.0 months [95% CI: (9.3 - 13.7)], respectively in the whole cohort and 10.8 [95% CI: (9.3 - 12.9)] and 15.0 [95% CI: (13.2 - 17.8)] for irradiated patients. Induction treatment led to corticosteroid dose reduction or cessation in 21 patients (54%). KPS improvement was observed in 38% of patients. Toxicity was mild with only 7/39 (18%) grade III-IV toxicity, including 1 digestive bleeding and 1 epistaxis. Conclusion.TMZ-BEV induction led to CTV reduction allowing for optimal chemoradiotherapy in a majority (62%) of patients for which radiotherapy was initially recused. A clinical benefit was obtained with improved KPS and a decrease in steroid dose.
Collapse
Affiliation(s)
- Ilan Darmon
- Department of Radiotherapy, University Hospital, Amiens, France
| | | | | | - Marie Blonski
- Department of Neuro-Oncology, University Hospital, Nancy, France
| | - Emilie Le Rhun
- Department of Neuro-Oncology, University Hospital, Lille, France
| | - Luc Taillandier
- Department of Neuro-Oncology, University Hospital, Nancy, France
| | | | | | - Stéphanie Trudel
- Laboratory of Molecular Oncology, University Hospital, Amiens, France
| | - Jean-Christophe Faivre
- Department of Radiotherapy, Centre Alexis Vautrin, Nancy, France.,Department of Radiotherapy, Hospital of Metz-Thionville, France
| | - Nicolas Blanchard
- Department of Radiotherapy, Clinique des Dentellières, Valenciennes, France
| | - Bruno Chauffert
- Department of Medical Oncology , University Hospital, Amiens, France
| | - Mathieu Boone
- Department of Medical Oncology , University Hospital, Amiens, France
| |
Collapse
|
146
|
Xu D, Jia F, Li G, Li H. Dosimetric comparison of intensity-modulated radiation therapy and volumetric-modulated arc therapy plans for the treatment of glioma using flattening filter-free and flattening filter modes. Oncol Lett 2017; 13:3451-3456. [PMID: 28521451 PMCID: PMC5431317 DOI: 10.3892/ol.2017.5883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/23/2017] [Indexed: 11/17/2022] Open
Abstract
In the present study, the dose verification between 6X and 6X flattening filter-free (FFF) in intensity-modulated radiation therapy (IMRT) and volumetric modulated arc therapy (VMAT) was compared, and the advantages and disadvantages of different radiotherapy plans were evaluated. All four plans achieved comparable heterogeneity and conformity indices. For frontal tumor, VMAT demonstrated more improved sparing of the brainstem compared with the IMRT (P=0.045); while in the model of FFF, the Dmax of eye lens was significantly reduced by 16–21% (P<0.001). The organs at risk (OARs) in the temporal lobe tumor were spared well in the IMRT plan. With the removal of FF, the low-dose volume for both tumor locations was significantly reduced (P<0.05). By contrast, there was no significant difference in monitor units (MUs) with FFF, but the MUs were significantly reduced in the VMAT plan (P<0.001). Regarding the protection of OARs, FFF appeared to be superior compared with FF. For the frontal glioma, the VMAT plan had more advantages, and for temporal lobe tumor, dynamic IMRT was more appropriate. The VMAT plan reduces the low-dose volume of normal brain tissues and the MUs. While the removal of FF may increase the dose rate, the shortened treatment delivery time may improve the accuracy of treatment due to intra-fractional patient motion.
Collapse
|
147
|
Central nervous system gliomas. Crit Rev Oncol Hematol 2017; 113:213-234. [DOI: 10.1016/j.critrevonc.2017.03.021] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 03/16/2017] [Accepted: 03/20/2017] [Indexed: 12/22/2022] Open
|
148
|
Convection-enhanced delivery of a hydrophilic nitrosourea ameliorates deficits and suppresses tumor growth in experimental spinal cord glioma models. Acta Neurochir (Wien) 2017; 159:939-946. [PMID: 28247160 DOI: 10.1007/s00701-017-3123-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/16/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Convection-enhanced delivery (CED) is a technique allowing local infusion of therapeutic agents into the central nervous system, circumventing the blood-brain or spinal cord barrier. OBJECTIVE To evaluate the utility of nimustine hydrochloride (ACNU) CED in controlling tumor progression in an experimental spinal cord glioma model. METHODS Toxicity studies were performed in 42 rats following the administration of 4 μl of ACNU CED into the mid-thoracic spinal cord at concentrations ranging from 0.1 to 10 mg/ml. Behavioral analyses and histological evaluations were performed to assess ACNU toxicity in the spinal cord. A survival study was performed in 32 rats following the implantation of 9 L cells into the T8 spinal cord. Seven days after the implantation, rats were assigned to four groups: ACNU CED (0.25 mg/ml; n = 8); ACNU intravenous (i.v.) (0.4 mg; n = 8); saline CED (n = 8); saline i.v. (n = 8). Hind limb movements were evaluated daily in all rats for 21 days. Tumor sizes were measured histologically. RESULTS The maximum tolerated ACNU concentration was 0.25 mg/ml. Preservation of hind limb motor function and tumor growth suppression was observed in the ACNU CED (0.25 mg/ml) and ACNU i.v. groups. Antitumor effects were more prominent in the ACNU CED group especially in behavioral analyses (P < 0.05; log-rank test). CONCLUSIONS ACNU CED had efficacy in controlling tumor growth and preserving neurological function in an experimental spinal cord tumor model. ACNU CED can be a viable treatment option for spinal cord high-grade glioma.
Collapse
|
149
|
Management of GBM: a problem of local recurrence. J Neurooncol 2017; 134:487-493. [PMID: 28378194 DOI: 10.1007/s11060-016-2347-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/23/2016] [Indexed: 01/22/2023]
Abstract
Forty years ago, adjuvant treatment of patients with GBM using fractionated radiotherapy following surgery was shown to substantially improve survival compared to surgery alone. However, even with the addition of temozolomide to radiotherapy, overall survival is quite limited and local failure remains a fundamental problem, despite multiple attempts to increase dose to the tumor target. This review presents the historical background and clinical rationale leading to the current standard of care consisting of 60 Gy total dose in 2 Gy fractions to the MRI-defined targets in younger, high performance status patients and more hypofractionated regimens in elderly and/or debilitated patients. Particle therapies offer the potential to increase local control while reducing dose and, potentially, long-term neurocognitive toxicity. However, improvements in systemic therapies for GBM will need to be implemented before the full benefits of improved local control can be realized.
Collapse
|
150
|
Archer TC, Mahoney EL, Pomeroy SL. Medulloblastoma: Molecular Classification-Based Personal Therapeutics. Neurotherapeutics 2017; 14:265-273. [PMID: 28386677 PMCID: PMC5398996 DOI: 10.1007/s13311-017-0526-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer genomics have revealed 4 distinct subgroups of medulloblastomas, each with unique transcription profiles, DNA alterations and clinical outcome. Molecular classification of medulloblastomas improves predictions of clinical outcome, allowing more accurate matching of intensity of conventional treatments with chemotherapy and radiation to overall prognosis and setting the stage for the introduction of targeted therapies.
Collapse
Affiliation(s)
- Tenley C Archer
- Department of Neurology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | | | - Scott L Pomeroy
- Department of Neurology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|