Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 9/2017

01-09-2017 | Epilepsy (CW Bazil, Section Editor)

Brain Tumor-Related Epilepsy: a Current Review of the Etiologic Basis and Diagnostic and Treatment Approaches

Author: Jeffrey M. Politsky

Published in: Current Neurology and Neuroscience Reports | Issue 9/2017

Login to get access

Abstract

Purpose of Review

The relationship of brain tumors and seizures remains poorly understood. This article is an overview of the epidemiology, etiology, and diagnostic and treatment approaches of tumor-related epilepsy primarily with regard to glial-based tumors, the most commonly considered brain tumor in this field.

Recent Findings

Over the past many years, several novel etiologic mechanisms to explain how tumors induce seizures have been developed, which this article reviews, including the roles of glutamate-induced excitotoxicity, matrix metalloproteinases, isocitrate dehydrogenase, methylguanine methyltransferase, and functional network connectivity. As well, diagnostic and treatment approaches vary considerably. This article summarizes the evidence and provides the rationale for a reconsideration of how we deliver pre-operative, peri-operative, and post-operative care to these patients.

Summary

Patients with brain tumors and epilepsy are a very challenging subgroup of patients, which necessitates not just a thorough understanding of the current principles regarding tumor-related epilepsy but also the development of collaborative research to advance our knowledge even further, and a concerted effort to develop a standardized, multi-disciplinary clinical approach to improve the care of these patients.
Literature
1.
go back to reference Cascino GD. Epilepsy and brain tumors: implications for treatment. Epilepsia. 1990;31(suppl):S37–44.PubMedCrossRef Cascino GD. Epilepsy and brain tumors: implications for treatment. Epilepsia. 1990;31(suppl):S37–44.PubMedCrossRef
2.
go back to reference Pace A, Bove L, Innocenti P, et al. Epilepsy and gliomas: incidence and treatment in 119 patients. J Exper Clin Can Res. 1998;17:479–82. Pace A, Bove L, Innocenti P, et al. Epilepsy and gliomas: incidence and treatment in 119 patients. J Exper Clin Can Res. 1998;17:479–82.
3.
go back to reference Lynam L, Lyons M, Drzkowski J, Sirven J, Noe K, Zimmerman R, et al. Frequency of seizures in patients with newly diagnosed brain tumors: a retrospective review. Clin Neurol Neurosurg. 2007;109:634–8. Lynam L, Lyons M, Drzkowski J, Sirven J, Noe K, Zimmerman R, et al. Frequency of seizures in patients with newly diagnosed brain tumors: a retrospective review. Clin Neurol Neurosurg. 2007;109:634–8.
4.
go back to reference Lote K, Stenwig AE, Skullerud K, Hirschber H. Prevalence and prognositic significance of epilepsy in patients with gliomas. Eur J Cancer. 1998;34:98–102.PubMedCrossRef Lote K, Stenwig AE, Skullerud K, Hirschber H. Prevalence and prognositic significance of epilepsy in patients with gliomas. Eur J Cancer. 1998;34:98–102.PubMedCrossRef
5.
go back to reference Olson JD, Riedel E, DeAngelis LM. Long-term outcome of low-grade oligodendroglioma and mixed glioma. Neurology. 2000;54:1442–8.PubMedCrossRef Olson JD, Riedel E, DeAngelis LM. Long-term outcome of low-grade oligodendroglioma and mixed glioma. Neurology. 2000;54:1442–8.PubMedCrossRef
6.
go back to reference Lebrun C, Fontaine D, Ramaioli A, Vandenbos F, Chanalet S, Lonjon M, et al., The Nice Brain Tumor Study Group. Long-term outcome of oligodendrogliomas. Neurology. 2004;62:1783–7.PubMedCrossRef Lebrun C, Fontaine D, Ramaioli A, Vandenbos F, Chanalet S, Lonjon M, et al., The Nice Brain Tumor Study Group. Long-term outcome of oligodendrogliomas. Neurology. 2004;62:1783–7.PubMedCrossRef
8.
go back to reference Fernandez-Concepcion O, Gomez-Garcia A, Bonet-Gorvea M. Cerebral tumors as a cause of late onset epilepsy. Rev Neurol. 1996;29:1142–6. Fernandez-Concepcion O, Gomez-Garcia A, Bonet-Gorvea M. Cerebral tumors as a cause of late onset epilepsy. Rev Neurol. 1996;29:1142–6.
9.
go back to reference Penfield W, Erickson TI. Relation of intracranial tumors and symptomatic epilepsy. Arch Neurol Psych. 1940;44:300–15.CrossRef Penfield W, Erickson TI. Relation of intracranial tumors and symptomatic epilepsy. Arch Neurol Psych. 1940;44:300–15.CrossRef
10.
go back to reference Van Breemen M, Rijsman R, Taphoorn M, Walchenbach R, Zwinkels H, Vecht C. Efficacy of anti-epileptic drugs in patients with gliomas and seizures. J Neurol. 2009;256:1519–26.PubMedCrossRef Van Breemen M, Rijsman R, Taphoorn M, Walchenbach R, Zwinkels H, Vecht C. Efficacy of anti-epileptic drugs in patients with gliomas and seizures. J Neurol. 2009;256:1519–26.PubMedCrossRef
11.
go back to reference Shady JA, Black PM, Kupsky WJ, et al. Seizures in children with supratentorial astroglial neoplasms. Pediatr Neurosurg. 1994;21:23–30.PubMedCrossRef Shady JA, Black PM, Kupsky WJ, et al. Seizures in children with supratentorial astroglial neoplasms. Pediatr Neurosurg. 1994;21:23–30.PubMedCrossRef
12.
go back to reference • Pallud J, Audreau E, Bionski M, Sanai N, Bauchet L, Fontaine D, et al. Epileptic seizures in diffuse low-grade gliomas in adults. Brain. 2014;137:449–62. This is a retrospective review that examines the natural course of diffuse low-grade glioma and seizures. PubMedCrossRef • Pallud J, Audreau E, Bionski M, Sanai N, Bauchet L, Fontaine D, et al. Epileptic seizures in diffuse low-grade gliomas in adults. Brain. 2014;137:449–62. This is a retrospective review that examines the natural course of diffuse low-grade glioma and seizures. PubMedCrossRef
13.
go back to reference Shamji M, Fric-Shamji E, Benoit B. Brain tumors and epilepsy: pathophysiology of peritumoral change. Neurosurg Rev. 2009;32:275–85.PubMedCrossRef Shamji M, Fric-Shamji E, Benoit B. Brain tumors and epilepsy: pathophysiology of peritumoral change. Neurosurg Rev. 2009;32:275–85.PubMedCrossRef
14.
go back to reference Wolf H, Roos D, Blumcke I, Pietsch T, Wiestler O. Perilesional neurochemical changes in focal epilepsies. Acta Neuropathol (Berl). 1996;91:376–84.CrossRef Wolf H, Roos D, Blumcke I, Pietsch T, Wiestler O. Perilesional neurochemical changes in focal epilepsies. Acta Neuropathol (Berl). 1996;91:376–84.CrossRef
15.
go back to reference Kohling R, Senner V, Paulus W, Speckmann E. Epileptiform activity preferentially arises outside tumor invasion zone in glioma xeno-transplants. Neurobiol Dis. 2006;22:64–75.PubMedCrossRef Kohling R, Senner V, Paulus W, Speckmann E. Epileptiform activity preferentially arises outside tumor invasion zone in glioma xeno-transplants. Neurobiol Dis. 2006;22:64–75.PubMedCrossRef
16.
go back to reference Rothman S. The neurotoxicity of excitatory amino acids is produced by passive chloride influx. J Neurosci. 1985;5:1483–9.PubMed Rothman S. The neurotoxicity of excitatory amino acids is produced by passive chloride influx. J Neurosci. 1985;5:1483–9.PubMed
17.
go back to reference Choi D. Ionic dependence of glutamate neurotoxicity. J Neurosci. 1987;7:369–79.PubMed Choi D. Ionic dependence of glutamate neurotoxicity. J Neurosci. 1987;7:369–79.PubMed
18.
go back to reference Meldrum B. The role of glutamate in epilepsy and other cns disorders. Neurol. 1994;44(Suppl 8):S14–23. Meldrum B. The role of glutamate in epilepsy and other cns disorders. Neurol. 1994;44(Suppl 8):S14–23.
19.
go back to reference Rothstein J, Dykes-Hoberg M, Pardo C, Bristol L, Jin L, Kuncl R, et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996;16:675–86.PubMedCrossRef Rothstein J, Dykes-Hoberg M, Pardo C, Bristol L, Jin L, Kuncl R, et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996;16:675–86.PubMedCrossRef
20.
go back to reference Tanaka K, Watase K, Manabe T, Yamada K, Watanabe M, Takahashi K, et al. Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter glt-1. Science. 1997;276:1702.CrossRef Tanaka K, Watase K, Manabe T, Yamada K, Watanabe M, Takahashi K, et al. Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter glt-1. Science. 1997;276:1702.CrossRef
21.
go back to reference Chen Y, Swanson R. The glutamate transporters EAAT2 and EAAT3 mediate cysteine uptake in cortical neuron cultures. J Neurochem. 2003;84:1332–9.PubMedCrossRef Chen Y, Swanson R. The glutamate transporters EAAT2 and EAAT3 mediate cysteine uptake in cortical neuron cultures. J Neurochem. 2003;84:1332–9.PubMedCrossRef
22.
go back to reference Lewerenz J, Hewett S, Huang Y, Lambros M, Gout P, Kalivas P, et al. The cysteine/glutamate antiporter system xc- in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18:522–55.PubMedPubMedCentralCrossRef Lewerenz J, Hewett S, Huang Y, Lambros M, Gout P, Kalivas P, et al. The cysteine/glutamate antiporter system xc- in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18:522–55.PubMedPubMedCentralCrossRef
24.
go back to reference Wang J, Erickson J, Fuji R, Ramachandran S, Gao P, Dinavah R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19.PubMedPubMedCentralCrossRef Wang J, Erickson J, Fuji R, Ramachandran S, Gao P, Dinavah R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19.PubMedPubMedCentralCrossRef
25.
go back to reference Hensely C, Wasti A, DeBeradinis R. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J Clin Invest. 2013;123:3678–84.CrossRef Hensely C, Wasti A, DeBeradinis R. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J Clin Invest. 2013;123:3678–84.CrossRef
26.
go back to reference Rosati A, Marconi S, Pollo B, Tomassini A, Lovato L, Maderna E, et al. Epilepsy in glioblastoma multiforme: correlation with glutamine synthetase levels. J Neuro-Oncol. 2009;93:319–24.CrossRef Rosati A, Marconi S, Pollo B, Tomassini A, Lovato L, Maderna E, et al. Epilepsy in glioblastoma multiforme: correlation with glutamine synthetase levels. J Neuro-Oncol. 2009;93:319–24.CrossRef
27.
go back to reference Rosati A, Polari P, Todeschini A, Cominelli M, Medicina D, Cenzato M, et al. Glutamine synthetase expression as a valuable marker of epilepsy and longer survival newly diagnosed glioblastoma multiforme. Neuro-Oncology. 2013;15:618–25.PubMedPubMedCentralCrossRef Rosati A, Polari P, Todeschini A, Cominelli M, Medicina D, Cenzato M, et al. Glutamine synthetase expression as a valuable marker of epilepsy and longer survival newly diagnosed glioblastoma multiforme. Neuro-Oncology. 2013;15:618–25.PubMedPubMedCentralCrossRef
28.
go back to reference Ye Z, Sontheimer H. Glioma cells release excitotoxic concentrations of glutamate. Cancer Res. 1999;59:4383–91.PubMed Ye Z, Sontheimer H. Glioma cells release excitotoxic concentrations of glutamate. Cancer Res. 1999;59:4383–91.PubMed
29.
30.
go back to reference Marcus H, Carpenter K, Prie S, Hutchinson P. In vivo assessment of high-grade glioma biochemistry using microdialysis: a study of energy-related molecules, growth factors, and cytokines. J Neuro-Oncol. 2010;97:11–23.CrossRef Marcus H, Carpenter K, Prie S, Hutchinson P. In vivo assessment of high-grade glioma biochemistry using microdialysis: a study of energy-related molecules, growth factors, and cytokines. J Neuro-Oncol. 2010;97:11–23.CrossRef
31.
go back to reference Yuen T, Morokoff A, Bjorksten A, D’Abaco G, Paradiso L, Finch S, et al. Glutamate is associated with a higher risk of seizures in patients with gliomas. Neurology. 2012;79:883–9.PubMedCrossRef Yuen T, Morokoff A, Bjorksten A, D’Abaco G, Paradiso L, Finch S, et al. Glutamate is associated with a higher risk of seizures in patients with gliomas. Neurology. 2012;79:883–9.PubMedCrossRef
32.
go back to reference • Buckingham S, Campbell S, Haas B, Montana V, Robel S, Ogunrinu T, et al. Glutamate release by primary brain tumors induces epileptic activity. Nat Med. 2012;17:1269–74. This paper from Sontheimer’s lab discusses the pathophysiologic effects of glutamate release from gliomas and the role of glutamate in tumor-related epilepsy. • Buckingham S, Campbell S, Haas B, Montana V, Robel S, Ogunrinu T, et al. Glutamate release by primary brain tumors induces epileptic activity. Nat Med. 2012;17:1269–74. This paper from Sontheimer’s lab discusses the pathophysiologic effects of glutamate release from gliomas and the role of glutamate in tumor-related epilepsy.
34.
go back to reference Phillips H, Kharbanda S, Chen R, Forrest W, Soriano R, Wu T, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157–73.PubMedCrossRef Phillips H, Kharbanda S, Chen R, Forrest W, Soriano R, Wu T, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157–73.PubMedCrossRef
35.
go back to reference Verhaak R, Hoadley K, Purdom E, Wang V, Qi Y, Wilderson M, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, and NF1. Cancer Cell. 2010;17:510–22.PubMedPubMedCentralCrossRef Verhaak R, Hoadley K, Purdom E, Wang V, Qi Y, Wilderson M, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, and NF1. Cancer Cell. 2010;17:510–22.PubMedPubMedCentralCrossRef
36.
go back to reference Berntsson S, Malmer B, Bondy M, Qu M, Smits A. Tumor-associated epilepsy and glioma: are there common genetic pathways? Acta Oncol. 2009;48:955–63.PubMedCrossRef Berntsson S, Malmer B, Bondy M, Qu M, Smits A. Tumor-associated epilepsy and glioma: are there common genetic pathways? Acta Oncol. 2009;48:955–63.PubMedCrossRef
37.
go back to reference Thom M, Blumcke I, Aronica E. Long-term epilepsy-associated tumors. Brian Pathol. 2012;22:350–79.CrossRef Thom M, Blumcke I, Aronica E. Long-term epilepsy-associated tumors. Brian Pathol. 2012;22:350–79.CrossRef
38.
go back to reference Esteller M, Garcia-Foncillas J, Andion E, Goodman S, Hidalgo O, Vanaclocha V, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343:1350–4. Esteller M, Garcia-Foncillas J, Andion E, Goodman S, Hidalgo O, Vanaclocha V, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343:1350–4.
39.
go back to reference Stupp R, Hegi M, Mason W, van den Bent M, Taphoorn M, Janzer R, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:459–66.PubMedCrossRef Stupp R, Hegi M, Mason W, van den Bent M, Taphoorn M, Janzer R, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:459–66.PubMedCrossRef
40.
go back to reference Kreth S, Thon N, Eigenbrod S, Lutz J, Ledderose C, Egensperger R, et al. O6-methylguanine-DNA methyltransferase (MGMT) mRNA expression predicts outcome in malignant glioma independent of MGMT promoter methylation. PLoS One. 2011;6(2):e17156.PubMedPubMedCentralCrossRef Kreth S, Thon N, Eigenbrod S, Lutz J, Ledderose C, Egensperger R, et al. O6-methylguanine-DNA methyltransferase (MGMT) mRNA expression predicts outcome in malignant glioma independent of MGMT promoter methylation. PLoS One. 2011;6(2):e17156.PubMedPubMedCentralCrossRef
41.
go back to reference Zhang K, Wang X, Zhou B, Zhang L. The prognostic value of MGMT promoter methylation in glioblastoma multiforme: a meta-analysis. Familial Cancer. 2013;12:449–58.PubMedCrossRef Zhang K, Wang X, Zhou B, Zhang L. The prognostic value of MGMT promoter methylation in glioblastoma multiforme: a meta-analysis. Familial Cancer. 2013;12:449–58.PubMedCrossRef
42.
go back to reference Kobow K, Kaspi A, Harikrishnan K, Kiese K, Ziemann M, Khurana I, et al. Deep sequencing reveals increased DNA methylation in chronic rat epilepsy. Acta Neuropathol. 2013a;126:741–56.PubMedPubMedCentralCrossRef Kobow K, Kaspi A, Harikrishnan K, Kiese K, Ziemann M, Khurana I, et al. Deep sequencing reveals increased DNA methylation in chronic rat epilepsy. Acta Neuropathol. 2013a;126:741–56.PubMedPubMedCentralCrossRef
43.
go back to reference Kobow K, El-Osta A, Blumcke I. The methylation hypothesis of pharmacoresistance in epilepsy. Epilepsia. 2013b;54(Suppl 2):41–7.PubMedCrossRef Kobow K, El-Osta A, Blumcke I. The methylation hypothesis of pharmacoresistance in epilepsy. Epilepsia. 2013b;54(Suppl 2):41–7.PubMedCrossRef
44.
go back to reference Nedivi E, Hervoni D, Naot D, Israell D, Citri Y. Numerous candidate plasticity-related genes revealed by differential cDNA cloning. Nature. 1993;363:718–22.PubMedCrossRef Nedivi E, Hervoni D, Naot D, Israell D, Citri Y. Numerous candidate plasticity-related genes revealed by differential cDNA cloning. Nature. 1993;363:718–22.PubMedCrossRef
45.
go back to reference Rivera S, Tremblay E, Timsit S, Canals O, Ben-Ari Y, Khrestchatisky M. Tissue inhibitor of metalloproteinases-1 (TIMP-1) is differentially induced in neruons and astrocytes after seizures: evidence or developmental, immediate early gene, and lesion response. J Neurosci. 1997;17:4223–35.PubMed Rivera S, Tremblay E, Timsit S, Canals O, Ben-Ari Y, Khrestchatisky M. Tissue inhibitor of metalloproteinases-1 (TIMP-1) is differentially induced in neruons and astrocytes after seizures: evidence or developmental, immediate early gene, and lesion response. J Neurosci. 1997;17:4223–35.PubMed
46.
go back to reference Wright J, Brown T, Harding J. Inhibition of matrix metalloproteinase-3 and -9 disrupts spatial memory. Neural Plasticity. 2007;2007:article ID 73813. Wright J, Brown T, Harding J. Inhibition of matrix metalloproteinase-3 and -9 disrupts spatial memory. Neural Plasticity. 2007;2007:article ID 73813.
47.
go back to reference Nagy V, Bozdagi O, Matynia A, Balcerzyk M, Okulski P, Dzwonek J, et al. Matrix metalloproteinase-9 I srequired for hippocampal late-phase long-term potentiation and memory. J Neurosci. 2006;26:1923–34. Nagy V, Bozdagi O, Matynia A, Balcerzyk M, Okulski P, Dzwonek J, et al. Matrix metalloproteinase-9 I srequired for hippocampal late-phase long-term potentiation and memory. J Neurosci. 2006;26:1923–34.
48.
go back to reference Okulski P, Jay T, Jaworski J, Duniec K, Dzwonek J, Konopacki F, et al. TIMP-1 abolishes MMP-9-dependent long-lasting long-term potentiation in the prefrontal cortex. Biol Psychiatry. 2007;62:359–62. Okulski P, Jay T, Jaworski J, Duniec K, Dzwonek J, Konopacki F, et al. TIMP-1 abolishes MMP-9-dependent long-lasting long-term potentiation in the prefrontal cortex. Biol Psychiatry. 2007;62:359–62.
49.
go back to reference Wright J & Harding J. Contributions of matrix metalloproteinases to neural plasticity, habituation, associative learning and drug addiction. Neural Plasticity. 2009;2009:article ID 579382. Wright J & Harding J. Contributions of matrix metalloproteinases to neural plasticity, habituation, associative learning and drug addiction. Neural Plasticity. 2009;2009:article ID 579382.
50.
go back to reference • Mizoguchi H, Yamada K, Nabeshima T. matrix metalloproteinases contribute to neuronal dysfunction in animal models of drug dependence, Alzheimer’s disease, and epilepsy. Biochemistry Research International. 2011;2011:article ID 681385. Matrix metalloproteinases is an enzyme family that has critical roles in several normal and disease states, highlighted in this paper. • Mizoguchi H, Yamada K, Nabeshima T. matrix metalloproteinases contribute to neuronal dysfunction in animal models of drug dependence, Alzheimer’s disease, and epilepsy. Biochemistry Research International. 2011;2011:article ID 681385. Matrix metalloproteinases is an enzyme family that has critical roles in several normal and disease states, highlighted in this paper.
51.
go back to reference Koyama R, Yamada M, Fujisawa S, Katoh-Semba R, Matsuki N, Ikegaya Y. Brain-derived neurotrophic factor induces hyperexcitable reentrant circuits in the dentate gyrus. J Neurosci. 2004;24:7215–24.PubMedCrossRef Koyama R, Yamada M, Fujisawa S, Katoh-Semba R, Matsuki N, Ikegaya Y. Brain-derived neurotrophic factor induces hyperexcitable reentrant circuits in the dentate gyrus. J Neurosci. 2004;24:7215–24.PubMedCrossRef
52.
go back to reference Wang M, Wang T, Liu S, Yoshida D, Teramoto A. The expressin of matrix metalloptroteinase-2 and -9 in human gliomas of different pathological grades. Brain Tumor Pathology. 2003;20:65–72.PubMedCrossRef Wang M, Wang T, Liu S, Yoshida D, Teramoto A. The expressin of matrix metalloptroteinase-2 and -9 in human gliomas of different pathological grades. Brain Tumor Pathology. 2003;20:65–72.PubMedCrossRef
53.
go back to reference Giannelli G, Falk-Marzillier J, Schiraldi O, Stetler-Stevenson W, Quaranta V. Induction of cell migration by matrix metalloproteinase-2 cleavage of laminin-5. Science. 1997;277:225–8.PubMedCrossRef Giannelli G, Falk-Marzillier J, Schiraldi O, Stetler-Stevenson W, Quaranta V. Induction of cell migration by matrix metalloproteinase-2 cleavage of laminin-5. Science. 1997;277:225–8.PubMedCrossRef
54.
go back to reference Dong J, Opresko L, Dempsey P, Lauffenburger D, Coffey R, Wiley H. Metalloproteinase-mediated ligand release regulates autocrine signaling through the epidermal growth factor receptor. Proc Natl Acad Sci. 1999;96:6235–40.PubMedPubMedCentralCrossRef Dong J, Opresko L, Dempsey P, Lauffenburger D, Coffey R, Wiley H. Metalloproteinase-mediated ligand release regulates autocrine signaling through the epidermal growth factor receptor. Proc Natl Acad Sci. 1999;96:6235–40.PubMedPubMedCentralCrossRef
55.
go back to reference Stettler-Stevenson W. Matrix metalloproteinases in angiogenesis: a moving target for therapeutic intervention. J Clin Investigation. 1999;103:1237–41.CrossRef Stettler-Stevenson W. Matrix metalloproteinases in angiogenesis: a moving target for therapeutic intervention. J Clin Investigation. 1999;103:1237–41.CrossRef
56.
go back to reference McCawley L, Matrisian L. Matrix metalloproteinases: they’re not just for matrix anymore! Curr Opin Cell Biol. 2001;13:534–40.PubMedCrossRef McCawley L, Matrisian L. Matrix metalloproteinases: they’re not just for matrix anymore! Curr Opin Cell Biol. 2001;13:534–40.PubMedCrossRef
57.
go back to reference Choe G, Park J, Jouben-Steele L, Kremen T, Liau L, Vinters H, et al. Active matrix metalloproteinase 9 expression is associated with primary glioblastoma subtype. Clin Cancer Res. 2002;8:2894–901. Choe G, Park J, Jouben-Steele L, Kremen T, Liau L, Vinters H, et al. Active matrix metalloproteinase 9 expression is associated with primary glioblastoma subtype. Clin Cancer Res. 2002;8:2894–901.
59.
go back to reference • Lubinas S, D’Abaco G, Moffat B, Gonzales M, Feleppa F, Nowell C, et al. IDH1 mutation is associated with seizures and protoplasmic subtype in patients with low-grade gliomas. Epilepsia. 2014;55:1438–43. This paper highlights the role of IDH1 mutation in tumor-related epilepsy. CrossRef • Lubinas S, D’Abaco G, Moffat B, Gonzales M, Feleppa F, Nowell C, et al. IDH1 mutation is associated with seizures and protoplasmic subtype in patients with low-grade gliomas. Epilepsia. 2014;55:1438–43. This paper highlights the role of IDH1 mutation in tumor-related epilepsy. CrossRef
60.
go back to reference Kolker S, Pawlak V, Ahlemeyer B, Okun J, Horster F, Mayatepek E, et al. NMDA receptor activation and respiratory chain complex V inhibition contribute to neurodegeneration in d-2-hydroxyglutaric aciduria. Eur J Neurosci. 2002;16:21–8.PubMedCrossRef Kolker S, Pawlak V, Ahlemeyer B, Okun J, Horster F, Mayatepek E, et al. NMDA receptor activation and respiratory chain complex V inhibition contribute to neurodegeneration in d-2-hydroxyglutaric aciduria. Eur J Neurosci. 2002;16:21–8.PubMedCrossRef
61.
63.
go back to reference De Groot M, Iyer A, Zurolo E, Anink J, Heimans J, Boison D, et al. Overexpression of ADK in human astrocytic tumors and pertumoral tissue is related to tumor-associated epilepsy. Epilepsia. 2012;53:58–66. De Groot M, Iyer A, Zurolo E, Anink J, Heimans J, Boison D, et al. Overexpression of ADK in human astrocytic tumors and pertumoral tissue is related to tumor-associated epilepsy. Epilepsia. 2012;53:58–66.
64.
go back to reference Watts DJ, Strogatz SH. Collective dynamics of ‘small-world’ networks. Nature. 1998;393:440–2.PubMedCrossRef Watts DJ, Strogatz SH. Collective dynamics of ‘small-world’ networks. Nature. 1998;393:440–2.PubMedCrossRef
65.
go back to reference Stam CJ. Functional connectivity patterns of human magnetoencephalographic recordings: a ‘small-world’ network? Neurosci Lett. 2004;355:25–8.PubMedCrossRef Stam CJ. Functional connectivity patterns of human magnetoencephalographic recordings: a ‘small-world’ network? Neurosci Lett. 2004;355:25–8.PubMedCrossRef
66.
go back to reference Salvador R, Suckling J, Schwarzbauer C, Bullmore E. Undirected graphs of frequency-dependent functional connectivity in whole brain networks. Philos Trans R Soc Lond Ser B Biol Sci. 2005;360:937–46.CrossRef Salvador R, Suckling J, Schwarzbauer C, Bullmore E. Undirected graphs of frequency-dependent functional connectivity in whole brain networks. Philos Trans R Soc Lond Ser B Biol Sci. 2005;360:937–46.CrossRef
67.
go back to reference He Y, Chen ZJ, Evans AC. Small-world anatomical networks in the human brain revealed by cortical thickness from MRI. Cereb Cortex. 2007;17:2407–19.PubMedCrossRef He Y, Chen ZJ, Evans AC. Small-world anatomical networks in the human brain revealed by cortical thickness from MRI. Cereb Cortex. 2007;17:2407–19.PubMedCrossRef
68.
go back to reference Bartolomei F, Bosma I, Klein M, Baayen JC, Reijneveld JC, Postma TJ, et al. How do brain tumors alter functional connectivity? A magnetoencephalography study. Ann Neurol. 2006a;59:128–38.PubMedCrossRef Bartolomei F, Bosma I, Klein M, Baayen JC, Reijneveld JC, Postma TJ, et al. How do brain tumors alter functional connectivity? A magnetoencephalography study. Ann Neurol. 2006a;59:128–38.PubMedCrossRef
69.
go back to reference Bartolomei F, Bosma I, Klein M, Baayen JC, Reijneveld JC, Postma TJ, et al. Disturbed functional connectivity in brain tumour patients: evaluation by graph analysis of synchronization matrices. Clin Neurophysiol. 2006b;117:2039–49.PubMedCrossRef Bartolomei F, Bosma I, Klein M, Baayen JC, Reijneveld JC, Postma TJ, et al. Disturbed functional connectivity in brain tumour patients: evaluation by graph analysis of synchronization matrices. Clin Neurophysiol. 2006b;117:2039–49.PubMedCrossRef
70.
go back to reference Bosma I, Douw L, Bartolomei F, Heimans J, van Dijk B, Postma T, et al. Synchronized brain activity and neurocognitive function in patients with low-grade glioma: a magnetoencephalography study. Neuro-Oncology. 2008;10:734–44.PubMedPubMedCentralCrossRef Bosma I, Douw L, Bartolomei F, Heimans J, van Dijk B, Postma T, et al. Synchronized brain activity and neurocognitive function in patients with low-grade glioma: a magnetoencephalography study. Neuro-Oncology. 2008;10:734–44.PubMedPubMedCentralCrossRef
71.
go back to reference Bosma I, Reijneveld J, Klein M, Douw L, van Dijk B, Heimans J, et al. Disturbed functional brain networks and neurocognitive function in lowgrade glioma patients: a graph theoretical analysis of resting-state MEG. Nonlinear Biomed Phys. 2009;3:9. Bosma I, Reijneveld J, Klein M, Douw L, van Dijk B, Heimans J, et al. Disturbed functional brain networks and neurocognitive function in lowgrade glioma patients: a graph theoretical analysis of resting-state MEG. Nonlinear Biomed Phys. 2009;3:9.
72.
go back to reference Douw L, de Groot M, van Dellen E, Heimans JJ, Ronner HE, Stam CJ. Reijneveld JC: ‘functional connectivity’ is a sensitive predictor of epilepsy diagnosis after the first seizure. PLoS One. 2010;5:e10839.PubMedPubMedCentralCrossRef Douw L, de Groot M, van Dellen E, Heimans JJ, Ronner HE, Stam CJ. Reijneveld JC: ‘functional connectivity’ is a sensitive predictor of epilepsy diagnosis after the first seizure. PLoS One. 2010;5:e10839.PubMedPubMedCentralCrossRef
73.
go back to reference Horstmann MT, Bialonski S, Noennig N, Mai H, Prusseit J, Wellmer J, et al. State dependent properties of epileptic brain networks: comparative graph-theoretical analyses of simultaneously recorded EEG and MEG. Clin Neurophysiol. 2010;121:172–85. Horstmann MT, Bialonski S, Noennig N, Mai H, Prusseit J, Wellmer J, et al. State dependent properties of epileptic brain networks: comparative graph-theoretical analyses of simultaneously recorded EEG and MEG. Clin Neurophysiol. 2010;121:172–85.
74.
go back to reference Liao W, Zhang Z, Pan Z, Mantini D, Ding J, Duan X, et al. Altered functional connectivity and small-world in mesial temporal lobe epilepsy. PLoS One. 2009;5:e8525. Liao W, Zhang Z, Pan Z, Mantini D, Ding J, Duan X, et al. Altered functional connectivity and small-world in mesial temporal lobe epilepsy. PLoS One. 2009;5:e8525.
75.
go back to reference van Dellen E, Douw L, Baayen JC, Heimans JJ, Ponten SC, Vandertop WP, et al. Long-term effects of temporal lobe epilepsy on local neural networks: a graph theoretical analysis of corticography recordings. PLoS One. 2009;4:e8081.PubMedPubMedCentralCrossRef van Dellen E, Douw L, Baayen JC, Heimans JJ, Ponten SC, Vandertop WP, et al. Long-term effects of temporal lobe epilepsy on local neural networks: a graph theoretical analysis of corticography recordings. PLoS One. 2009;4:e8081.PubMedPubMedCentralCrossRef
76.
go back to reference Douw L, van Dellen E, De Groot M, Heimans J, Klein M, Stam C, et al. Epilepsy is related to theta ban brain connectivity and network topology in brain tumor patients. BMC Neurosci. 2010;11:103. Douw L, van Dellen E, De Groot M, Heimans J, Klein M, Stam C, et al. Epilepsy is related to theta ban brain connectivity and network topology in brain tumor patients. BMC Neurosci. 2010;11:103.
77.
go back to reference • Glantz MJ, Cole BF, Forsyth PA, et al. Practice parameter: anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Neurology. 2000;54:1886–93. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. PubMedCrossRef • Glantz MJ, Cole BF, Forsyth PA, et al. Practice parameter: anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Neurology. 2000;54:1886–93. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. PubMedCrossRef
78.
go back to reference Sirven J, Wingerchuk D, Drazkowski J, Lyons M, Zimmerman R. Seizure prophylaxis in patients with brain tumors: a meta-analysis. Mayo Clin Proc. 2004;79:1489–94.PubMedCrossRef Sirven J, Wingerchuk D, Drazkowski J, Lyons M, Zimmerman R. Seizure prophylaxis in patients with brain tumors: a meta-analysis. Mayo Clin Proc. 2004;79:1489–94.PubMedCrossRef
79.
go back to reference • Tremont-Lukats I, Ratilal B, Armstrong T, Gilbert M. Antiepileptic drugs for preventing seizures in patients with brain tumors. Cochrane Database Syst Rev. 2008;2:CD004424. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. • Tremont-Lukats I, Ratilal B, Armstrong T, Gilbert M. Antiepileptic drugs for preventing seizures in patients with brain tumors. Cochrane Database Syst Rev. 2008;2:CD004424. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions.
80.
go back to reference Kong X, Guan J, Yang Y, Li Y, Ma W, Wang R. A meta-analysis: do prophylactic antiepileptic drugs in patients with brain tumors decrease the incidence of seizures? Clin Neurol Neurosurg. 2015;134:98–103.PubMedCrossRef Kong X, Guan J, Yang Y, Li Y, Ma W, Wang R. A meta-analysis: do prophylactic antiepileptic drugs in patients with brain tumors decrease the incidence of seizures? Clin Neurol Neurosurg. 2015;134:98–103.PubMedCrossRef
81.
go back to reference • Sayegh E, Fakurnejad S, Oh T, Bloch O, Parsa A. Anti-convulsant prophylaxis for brain tumor surgery: determining the current best available evidence. J Neurosurg. 2014;121:1139–47. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. PubMedCrossRef • Sayegh E, Fakurnejad S, Oh T, Bloch O, Parsa A. Anti-convulsant prophylaxis for brain tumor surgery: determining the current best available evidence. J Neurosurg. 2014;121:1139–47. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. PubMedCrossRef
82.
go back to reference Glantz M, Friedberg M, Cole B, et al. Double-blind, randomized, placebo-controlled trial of anticonvulsant prophylaxis in adults with newly diagnosed brain metastases. Proc Amer Soc Clin Oncol. 1994;13:176. Glantz M, Friedberg M, Cole B, et al. Double-blind, randomized, placebo-controlled trial of anticonvulsant prophylaxis in adults with newly diagnosed brain metastases. Proc Amer Soc Clin Oncol. 1994;13:176.
83.
go back to reference Glantz MJ, Cole BF, Friedberg MH, et al. A randomized, blinded, placebo-controlled trial of divalproex sodium prophylaxis in adults with newly diagnosed brain tumors. Neurology. 1996;46:985–91.PubMedCrossRef Glantz MJ, Cole BF, Friedberg MH, et al. A randomized, blinded, placebo-controlled trial of divalproex sodium prophylaxis in adults with newly diagnosed brain tumors. Neurology. 1996;46:985–91.PubMedCrossRef
84.
go back to reference • de Oliveira J, Santana I, Caires I, Caires-Lima I, Miranda V, Protasio B, et al. Antiepileptic drug prophylaxis in primary brain tumor patients: is current practice in agreement to the consensus. J Neuro-Oncol. 2014;120:399–403. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. CrossRef • de Oliveira J, Santana I, Caires I, Caires-Lima I, Miranda V, Protasio B, et al. Antiepileptic drug prophylaxis in primary brain tumor patients: is current practice in agreement to the consensus. J Neuro-Oncol. 2014;120:399–403. This sample of sequential publications represents, including meta-analyses, represents independent evaluations of the role of prophylactic anti-seizure drug therapy in brain tumor patients, all with similar conclusions. CrossRef
85.
go back to reference Wu A, Trinh V, Suki D, Graham S, Forman A, Weinberg J, et al. A prospective randomized trial of perioperative seizure prophylaxis in patients with intraparenchymal brain tumors. J Neurosurg. 2013;118:873–83.PubMedPubMedCentralCrossRef Wu A, Trinh V, Suki D, Graham S, Forman A, Weinberg J, et al. A prospective randomized trial of perioperative seizure prophylaxis in patients with intraparenchymal brain tumors. J Neurosurg. 2013;118:873–83.PubMedPubMedCentralCrossRef
86.
go back to reference Garbossa D, Panciani P, Angeleri R, Battaglia L, Tartara F, Ajello M, et al. A retrospective two-center study of antiepileptic prophylaxis in patients with surgically treated high-grade gliomas. Neurol India. 2013;61:131–7.PubMedCrossRef Garbossa D, Panciani P, Angeleri R, Battaglia L, Tartara F, Ajello M, et al. A retrospective two-center study of antiepileptic prophylaxis in patients with surgically treated high-grade gliomas. Neurol India. 2013;61:131–7.PubMedCrossRef
87.
go back to reference Ansari S, Bohnstedt B, Perkins S, Althouse S, Miller J. Efficacy of postoperative seizure prophylaxis in intra-axial brain tumor resections. J Neuro-Oncol. 2014;118:117–22.CrossRef Ansari S, Bohnstedt B, Perkins S, Althouse S, Miller J. Efficacy of postoperative seizure prophylaxis in intra-axial brain tumor resections. J Neuro-Oncol. 2014;118:117–22.CrossRef
88.
go back to reference Lee Y, Kim T, Bae S, Kim Y, Han J, Yun C, et al. Levetiracetam compared with valproic acid for the prevention of postoperative seizures after supratentorial tumor surgery: a retrospective chart review. CNS Drugs. 2013;27:753–9. Lee Y, Kim T, Bae S, Kim Y, Han J, Yun C, et al. Levetiracetam compared with valproic acid for the prevention of postoperative seizures after supratentorial tumor surgery: a retrospective chart review. CNS Drugs. 2013;27:753–9.
89.
go back to reference Siomin V, Angelov L, Li L, Vogelbaum M. Results of a survey of neurosurgical practice patterns regarding the prophylactic use of anti-epilepsy drugs in patients with brain tumors. J Neuro-Oncol. 2005;74:211–5.CrossRef Siomin V, Angelov L, Li L, Vogelbaum M. Results of a survey of neurosurgical practice patterns regarding the prophylactic use of anti-epilepsy drugs in patients with brain tumors. J Neuro-Oncol. 2005;74:211–5.CrossRef
90.
go back to reference Hildebrand J. Management of epileptic seizures. Curr Op Oncol. 2004;16:314–7.CrossRef Hildebrand J. Management of epileptic seizures. Curr Op Oncol. 2004;16:314–7.CrossRef
91.
go back to reference Aguiar D, Pazo R, Duran I, Terrasa J, Arrivi A, Manzano H, et al. Toxic epidermal necrolysis in patients receiving anticonvulsants and cranial irradiation: a risk to consider. J Neuro-Oncol. 2004;66:345–50.CrossRef Aguiar D, Pazo R, Duran I, Terrasa J, Arrivi A, Manzano H, et al. Toxic epidermal necrolysis in patients receiving anticonvulsants and cranial irradiation: a risk to consider. J Neuro-Oncol. 2004;66:345–50.CrossRef
92.
go back to reference Taberner-Bonastre M, Peralta-Munoz S, Boza F, Guma I, Padro J. Neutropenia secondary to exposure to levetiracetam. Tumori. 2015;101:e145–6.PubMed Taberner-Bonastre M, Peralta-Munoz S, Boza F, Guma I, Padro J. Neutropenia secondary to exposure to levetiracetam. Tumori. 2015;101:e145–6.PubMed
93.
go back to reference Bilgili S, Calka O, Karadag A, Burakgazi A. EMPACT syndrome. Cutan Ocular Toxicol. 2011;30:328–30.CrossRef Bilgili S, Calka O, Karadag A, Burakgazi A. EMPACT syndrome. Cutan Ocular Toxicol. 2011;30:328–30.CrossRef
94.
go back to reference Zachenhofer I, Donat M, Oberndorfer S, Roessler K. Perioperative levetiracetam for prevention of seizures in supratentorial brain tumor surgery. J Neuro-Oncol. 2011;101:101–6.CrossRef Zachenhofer I, Donat M, Oberndorfer S, Roessler K. Perioperative levetiracetam for prevention of seizures in supratentorial brain tumor surgery. J Neuro-Oncol. 2011;101:101–6.CrossRef
95.
go back to reference Kandil A, Dvorak T, Mignano J, Wu J, Zhu J. Multifocal Stevens-Johnson syndrome after concurrent phenytoin and cranial and thoracic radiation treatment, a case report. Radiat Oncol. 2010;5:49.PubMedPubMedCentralCrossRef Kandil A, Dvorak T, Mignano J, Wu J, Zhu J. Multifocal Stevens-Johnson syndrome after concurrent phenytoin and cranial and thoracic radiation treatment, a case report. Radiat Oncol. 2010;5:49.PubMedPubMedCentralCrossRef
96.
go back to reference Beswick T, Cohen J. Dose-related levetiracetam-induced reticulated drug eruption. J Drugs Dermatol. 2010;9:409–10.PubMed Beswick T, Cohen J. Dose-related levetiracetam-induced reticulated drug eruption. J Drugs Dermatol. 2010;9:409–10.PubMed
97.
go back to reference Metro G, Pino S, Pellegrini D, Sacerdoti G, Fabi A. Brain radiotherapy during treatment with anticonvulsant therapy as a trigger for toxic epidermal necrolysis. Anticancer Res. 2007;27:1167–9.PubMed Metro G, Pino S, Pellegrini D, Sacerdoti G, Fabi A. Brain radiotherapy during treatment with anticonvulsant therapy as a trigger for toxic epidermal necrolysis. Anticancer Res. 2007;27:1167–9.PubMed
98.
go back to reference • Lee C, Lai H, Chiu A, Chan S, Hsiao L, Lee S. The effects of antiepileptic drugs on the growth of glioblastoma cell lines. J Neuro-Oncol. 2016;127:445–53. The paper represents an interesting and novel approach that investigates the effects of multiple conventional anti-seizure drugs on the cellular growth of malignant gliomas. CrossRef • Lee C, Lai H, Chiu A, Chan S, Hsiao L, Lee S. The effects of antiepileptic drugs on the growth of glioblastoma cell lines. J Neuro-Oncol. 2016;127:445–53. The paper represents an interesting and novel approach that investigates the effects of multiple conventional anti-seizure drugs on the cellular growth of malignant gliomas. CrossRef
99.
go back to reference Gefroh-Grimes H, Gidal B. Antiepileptic drugs in patients with malignant brain tumor: beyond seizures and pharmacokinetics. Acta Scandinavica. 2016;133:4–16. Gefroh-Grimes H, Gidal B. Antiepileptic drugs in patients with malignant brain tumor: beyond seizures and pharmacokinetics. Acta Scandinavica. 2016;133:4–16.
100.
go back to reference Kerkhof M, Dielemans J, Van Breemen M, Zwinkels H, Walchenbach R, Taphoorn M, et al. Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro-Oncology. 2013;15:961–7. Kerkhof M, Dielemans J, Van Breemen M, Zwinkels H, Walchenbach R, Taphoorn M, et al. Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro-Oncology. 2013;15:961–7.
101.
go back to reference Happold C, Gorlia T, Chinot O, Gilbert M, Nabors L, Wick W, et al. Does valproic acid or levetiracetam improve survival in glioblastoma? A pooled analysis of prospective clinical trials in newly diagnosed glioblastoma. J Clin Oncol. 2016;34:731–9.PubMedPubMedCentralCrossRef Happold C, Gorlia T, Chinot O, Gilbert M, Nabors L, Wick W, et al. Does valproic acid or levetiracetam improve survival in glioblastoma? A pooled analysis of prospective clinical trials in newly diagnosed glioblastoma. J Clin Oncol. 2016;34:731–9.PubMedPubMedCentralCrossRef
102.
go back to reference Robe P, Martin D, Nguyen-Khac M, Artesi M, Deprez M, Albert A, et al. Early termination of ISRCTN45828668, a phase 1/2 prospective, randomized study of sulfasalazine for the treatment of progressing malignant gliomas in adults. BMC Cancer. 2009;9:372–9.PubMedPubMedCentralCrossRef Robe P, Martin D, Nguyen-Khac M, Artesi M, Deprez M, Albert A, et al. Early termination of ISRCTN45828668, a phase 1/2 prospective, randomized study of sulfasalazine for the treatment of progressing malignant gliomas in adults. BMC Cancer. 2009;9:372–9.PubMedPubMedCentralCrossRef
103.
go back to reference Grossman S, Ye X, Chamberlain M, Mikkelsen T, Batchelor T, Desideri S, et al. Talampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trial. J Clin Oncol. 2009;27:4155–61. Grossman S, Ye X, Chamberlain M, Mikkelsen T, Batchelor T, Desideri S, et al. Talampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trial. J Clin Oncol. 2009;27:4155–61.
104.
go back to reference • Amar S, Fields G. Potential clinical implications of recent MMP inhibitor design strategies. Expert Rev Proteomics. 2015;12:445–7. Matrix metalloproteinases is an enzyme family that has critical roles in several normal and disease states, highlighted in this paper. PubMedPubMedCentralCrossRef • Amar S, Fields G. Potential clinical implications of recent MMP inhibitor design strategies. Expert Rev Proteomics. 2015;12:445–7. Matrix metalloproteinases is an enzyme family that has critical roles in several normal and disease states, highlighted in this paper. PubMedPubMedCentralCrossRef
105.
go back to reference Ikonomidou Chrysanthy. Matrix metalloproteinases and epileptogenesis. Mol Cell Pediatr. 2014;1:1–6.CrossRef Ikonomidou Chrysanthy. Matrix metalloproteinases and epileptogenesis. Mol Cell Pediatr. 2014;1:1–6.CrossRef
106.
go back to reference Cathcart J, Pulkoski-Gross A, Cao J. Targeting matrix metalloproteinases in cancer: bringing new life to old ideas. Genes & Diseases. 2015;2:26–34.CrossRef Cathcart J, Pulkoski-Gross A, Cao J. Targeting matrix metalloproteinases in cancer: bringing new life to old ideas. Genes & Diseases. 2015;2:26–34.CrossRef
107.
go back to reference Stellas D, Patsavoudi E. Inhibiting matrix metalloproteinases, an old story with new potentials for cancer treatment. Anti Cancer Agents Med Chem. 2012;12:707–17.CrossRef Stellas D, Patsavoudi E. Inhibiting matrix metalloproteinases, an old story with new potentials for cancer treatment. Anti Cancer Agents Med Chem. 2012;12:707–17.CrossRef
108.
go back to reference Kranendijk M, Salomons GS, Gibson K, Van Schaftingen E, Jakobs C, Struys E. A lymphoblast model for IDH2 gain-of-function activity in D-2-hydroxyglutaric aciduria type II: novel avenues for biochemical and therapeutic studies. Biochim Biophys Acta. 1812;2011:1380–4. Kranendijk M, Salomons GS, Gibson K, Van Schaftingen E, Jakobs C, Struys E. A lymphoblast model for IDH2 gain-of-function activity in D-2-hydroxyglutaric aciduria type II: novel avenues for biochemical and therapeutic studies. Biochim Biophys Acta. 1812;2011:1380–4.
109.
go back to reference Valadez J, Grover V, Carter M, Calcuttc M, Abiria S, Lundberg C, et al. Identification of Hedgehog pathway responsive glioblastomas by isocitrate dehydrogenase mutation. Cancer Lett. 2013;328:297–306. Valadez J, Grover V, Carter M, Calcuttc M, Abiria S, Lundberg C, et al. Identification of Hedgehog pathway responsive glioblastomas by isocitrate dehydrogenase mutation. Cancer Lett. 2013;328:297–306.
110.
go back to reference Fathi A, Abdel-Wahab O. Mutations in epigenetic modifiers in myeloid malignancies and the prospect of novel epigenetic-targeted therapy. Adv Hematol. 2012;2012:469592.PubMedCrossRef Fathi A, Abdel-Wahab O. Mutations in epigenetic modifiers in myeloid malignancies and the prospect of novel epigenetic-targeted therapy. Adv Hematol. 2012;2012:469592.PubMedCrossRef
113.
go back to reference Veech R, Chance B, Kashiwaya Y, Lardy H, Cahill G Jr. Ketone bodies, potential therapeutic uses. IUBMB Life. 2001;51:241–7.PubMedCrossRef Veech R, Chance B, Kashiwaya Y, Lardy H, Cahill G Jr. Ketone bodies, potential therapeutic uses. IUBMB Life. 2001;51:241–7.PubMedCrossRef
115.
go back to reference Seyfried TN, Mukherjee P. Targeting energy metabolism in brain cancer: review and hypothesis. Nutr Metab (Lond). 2005;2:30–8.CrossRef Seyfried TN, Mukherjee P. Targeting energy metabolism in brain cancer: review and hypothesis. Nutr Metab (Lond). 2005;2:30–8.CrossRef
116.
go back to reference Zhou WP, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, Seyfried TN. The calorically restricted ketongenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond). 2007;4:5.CrossRef Zhou WP, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, Seyfried TN. The calorically restricted ketongenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond). 2007;4:5.CrossRef
117.
go back to reference Maurer G, Brucker D, Bahr O, Harter P, Hattingen E, Walenta S, et al. Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale for ketogenic diet as experimental glioma therapy. BMC Cancer. 2011;11:315. Maurer G, Brucker D, Bahr O, Harter P, Hattingen E, Walenta S, et al. Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale for ketogenic diet as experimental glioma therapy. BMC Cancer. 2011;11:315.
118.
go back to reference Seyfreid TN, Kiebish MA, Marsh J, Shelton LM, Huysentruyt LC, Mukherjee P. Metabolic management of brain cancer. Biocheim Biophys Acta. 1807;2011:577–94. Seyfreid TN, Kiebish MA, Marsh J, Shelton LM, Huysentruyt LC, Mukherjee P. Metabolic management of brain cancer. Biocheim Biophys Acta. 1807;2011:577–94.
119.
go back to reference Seyfried TN. Cancer as a metabolic disease: on the origin management and prevention of cancer. INc, Hoboken, NJ: John Wiley and Sons; 2012.CrossRef Seyfried TN. Cancer as a metabolic disease: on the origin management and prevention of cancer. INc, Hoboken, NJ: John Wiley and Sons; 2012.CrossRef
121.
go back to reference Mukherjee P, El-Abbadi M, Kasperzyk J, Ranes M, Seyfried T. Dietary restriction reduces angiogenesis and growth in an orthoptic mouse brain tumour model. Br J Cancer. 2002;86:1615–21.PubMedPubMedCentralCrossRef Mukherjee P, El-Abbadi M, Kasperzyk J, Ranes M, Seyfried T. Dietary restriction reduces angiogenesis and growth in an orthoptic mouse brain tumour model. Br J Cancer. 2002;86:1615–21.PubMedPubMedCentralCrossRef
122.
go back to reference Mukherjee P, Abate L, Seyfried T. Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin Cancer Res. 2004;10:5622–9.PubMedCrossRef Mukherjee P, Abate L, Seyfried T. Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin Cancer Res. 2004;10:5622–9.PubMedCrossRef
123.
go back to reference Shelton L, Huysentruyt C, Mukherjee P, Seyfried T. Calorie restriction as an anti-invasive therapy for malignant brain cancer in the VM mouse. ASN Neuro. 2010;2:e00038.PubMedPubMedCentralCrossRef Shelton L, Huysentruyt C, Mukherjee P, Seyfried T. Calorie restriction as an anti-invasive therapy for malignant brain cancer in the VM mouse. ASN Neuro. 2010;2:e00038.PubMedPubMedCentralCrossRef
124.
go back to reference Jiang YS, Wang FR. Caloric restriction reduces edema and prolongs survival in a mouse glioma model. J Neruooncol. 2013;114:25–32.CrossRef Jiang YS, Wang FR. Caloric restriction reduces edema and prolongs survival in a mouse glioma model. J Neruooncol. 2013;114:25–32.CrossRef
125.
go back to reference Lee C, Raffaghello L, Brandhorst S, Safdie M, Bianchi G, Marin-Montalvo A, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4:124ra27.PubMedPubMedCentralCrossRef Lee C, Raffaghello L, Brandhorst S, Safdie M, Bianchi G, Marin-Montalvo A, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4:124ra27.PubMedPubMedCentralCrossRef
126.
go back to reference Scheck A, Abdelwahab M, Fenton K, Stafford P. The ketogenic diet for the treatment of glioma: insights from genetic profiling. Epilepsy Res. 2012;100:327–37.PubMedCrossRef Scheck A, Abdelwahab M, Fenton K, Stafford P. The ketogenic diet for the treatment of glioma: insights from genetic profiling. Epilepsy Res. 2012;100:327–37.PubMedCrossRef
127.
go back to reference • Woolf E, Scheck A. The ketogenic diet for treatment of malignant glioma. J Lipid Res. 2015;56:5–10. This paper reviews the effects of the ketogenic diet, a known therapy for refractory epilepsy, on malignant glioma progression. PubMedPubMedCentralCrossRef • Woolf E, Scheck A. The ketogenic diet for treatment of malignant glioma. J Lipid Res. 2015;56:5–10. This paper reviews the effects of the ketogenic diet, a known therapy for refractory epilepsy, on malignant glioma progression. PubMedPubMedCentralCrossRef
128.
go back to reference Abdelwahab M, Fenton K, Preul M, Rho J, Lynch A, Stafford P, et al. The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant gliomas. PLoS One. 2012;7:e36197. Abdelwahab M, Fenton K, Preul M, Rho J, Lynch A, Stafford P, et al. The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant gliomas. PLoS One. 2012;7:e36197.
129.
go back to reference Klein P. Mid-Atlantic Epilepsy Center (personal communication, 2017). Klein P. Mid-Atlantic Epilepsy Center (personal communication, 2017).
130.
131.
go back to reference • Rosenow F, Luders H. Presurgical evaluation of epilepsy. Brain. 2001;124:1683–700. This paper provides a thorough a review regarding the pre-surgical work-up in refractory epilepsy. PubMedCrossRef • Rosenow F, Luders H. Presurgical evaluation of epilepsy. Brain. 2001;124:1683–700. This paper provides a thorough a review regarding the pre-surgical work-up in refractory epilepsy. PubMedCrossRef
132.
go back to reference Winkler P. Extraoperative mapping. In: Duffau H, editor. Brain mapping: from neural basis of cognition to surgical applications. NY: Springer Wien. New York; 2011. p. 91–100.CrossRef Winkler P. Extraoperative mapping. In: Duffau H, editor. Brain mapping: from neural basis of cognition to surgical applications. NY: Springer Wien. New York; 2011. p. 91–100.CrossRef
133.
go back to reference Hamer H, Najm I, Mohamed A, Wyllie E. Interictal epileptiform discharges in temporal lobe epilepsy due to hippocampal sclerosis versus medial temporal lobe tumors. Epilepsia. 1999;40:1261–8.PubMedCrossRef Hamer H, Najm I, Mohamed A, Wyllie E. Interictal epileptiform discharges in temporal lobe epilepsy due to hippocampal sclerosis versus medial temporal lobe tumors. Epilepsia. 1999;40:1261–8.PubMedCrossRef
134.
go back to reference Duffau H. Brain mapping in tumors: intraoperative or extraoperative. Epilepsia. 2013;54(Suppl 9):79–83.PubMedCrossRef Duffau H. Brain mapping in tumors: intraoperative or extraoperative. Epilepsia. 2013;54(Suppl 9):79–83.PubMedCrossRef
135.
go back to reference Duffau H. The challenge to remove diffuse low grade gliomas while preserving brain functions. Acta Neurochir. 2012;154:569–74.PubMedCrossRef Duffau H. The challenge to remove diffuse low grade gliomas while preserving brain functions. Acta Neurochir. 2012;154:569–74.PubMedCrossRef
136.
go back to reference • Guenot M, Isnard J, Ryvlin P, FFischer C, Ostrowsky K, Mauguiere F, et al. Neruophysiological monitoring for epilepsy surgery: the Talairach SEEG method. Stereotact Funct Neurosurg. 2001;77:29–32. This paper provides a thorough a review regarding the pre-surgical work-up in refractory epilepsy. • Guenot M, Isnard J, Ryvlin P, FFischer C, Ostrowsky K, Mauguiere F, et al. Neruophysiological monitoring for epilepsy surgery: the Talairach SEEG method. Stereotact Funct Neurosurg. 2001;77:29–32. This paper provides a thorough a review regarding the pre-surgical work-up in refractory epilepsy.
137.
go back to reference Munari C, Hoffmann D, Francione S, Kahane P, Tassi L, Lo Russo G, et al. Stereo-electro-encephalography methodology: advantages and limits. Acta Neurol Scand Suppl. 1994;152:56–67. Munari C, Hoffmann D, Francione S, Kahane P, Tassi L, Lo Russo G, et al. Stereo-electro-encephalography methodology: advantages and limits. Acta Neurol Scand Suppl. 1994;152:56–67.
138.
go back to reference Talairach J, Bancaud J. Stereotaxic approach to epilepsy: methodology of anatomo-functional stereotactic investigations. Prog Neurol Surg. 1973;5:297–354.CrossRef Talairach J, Bancaud J. Stereotaxic approach to epilepsy: methodology of anatomo-functional stereotactic investigations. Prog Neurol Surg. 1973;5:297–354.CrossRef
139.
go back to reference Hamer H, Morris H, Mascha E, Karafa M, Bingaman W, Bej M, et al. Complications of invasive video-EEG monitoring with subdural grid electrodes. Neurology. 2002;58:97–103. Hamer H, Morris H, Mascha E, Karafa M, Bingaman W, Bej M, et al. Complications of invasive video-EEG monitoring with subdural grid electrodes. Neurology. 2002;58:97–103.
140.
go back to reference Onal C, Otsubo H, Araki T, Chitoku S, Ochi A, Weiss S, et al. Complications of invasive subdural grid monitoring in children with epilepsy. J Neurosurg. 2003;98:1017–26. Onal C, Otsubo H, Araki T, Chitoku S, Ochi A, Weiss S, et al. Complications of invasive subdural grid monitoring in children with epilepsy. J Neurosurg. 2003;98:1017–26.
141.
go back to reference Johnston J Jr, Mangano F, Ojemann J, Park T, Trevathan E, Smyth M. Complications of invasive subdrual electrode Monitorin at St. Louis Children’s Hospital, 1994-2005. J Neurosurg. 2006;105:343–7.PubMed Johnston J Jr, Mangano F, Ojemann J, Park T, Trevathan E, Smyth M. Complications of invasive subdrual electrode Monitorin at St. Louis Children’s Hospital, 1994-2005. J Neurosurg. 2006;105:343–7.PubMed
142.
go back to reference Wong C, Birkett J, Byth K, Dexter M, Somerville E, Gill D, et al. Risk factors for complications during intracranial electrode recording in presurgical evaluation of drug resistant partial epilepsy. Acta Neurochir. 2009;151:37–50. Wong C, Birkett J, Byth K, Dexter M, Somerville E, Gill D, et al. Risk factors for complications during intracranial electrode recording in presurgical evaluation of drug resistant partial epilepsy. Acta Neurochir. 2009;151:37–50.
143.
go back to reference Wellmer J, von der Groeben F, Klarmann U, Weber C, Elger C, Urbach H, et al. Risks and benefits of invasive epilepsy surgery workup with implanted subdural and depth electrodes. Epilepsia. 2012;53:1322–32. Wellmer J, von der Groeben F, Klarmann U, Weber C, Elger C, Urbach H, et al. Risks and benefits of invasive epilepsy surgery workup with implanted subdural and depth electrodes. Epilepsia. 2012;53:1322–32.
144.
go back to reference Bekelis K, Radwan T, Desai K, Moses Z, Thadani V, Jobst B, et al. Subdural interhemispheric grid electrodes for intracranial epilepsymonitoring: feasibility, safety, and utility: clinical article. J Neurosurg. 2012;117:1182–8.PubMedCrossRef Bekelis K, Radwan T, Desai K, Moses Z, Thadani V, Jobst B, et al. Subdural interhemispheric grid electrodes for intracranial epilepsymonitoring: feasibility, safety, and utility: clinical article. J Neurosurg. 2012;117:1182–8.PubMedCrossRef
145.
go back to reference Vale F, Pollock G, Dionisio J, Benbadis S, Tatum W. Outcome and complications of chronically implanted subdural electrodes for the treatment of medically resistant epilepsy. Clin Neurol Neurosurg. 2013;115:985–90.PubMedCrossRef Vale F, Pollock G, Dionisio J, Benbadis S, Tatum W. Outcome and complications of chronically implanted subdural electrodes for the treatment of medically resistant epilepsy. Clin Neurol Neurosurg. 2013;115:985–90.PubMedCrossRef
146.
go back to reference Tellez-Zenteno J, Hernandez-Ronquillo L, Moien-Afshari F, Wiebe S. Surgical outcomes in lesional and non-lesional epilepsy: a systematic review and meta-analysis. Epilepsy Res. 2010;89:310–8.PubMedCrossRef Tellez-Zenteno J, Hernandez-Ronquillo L, Moien-Afshari F, Wiebe S. Surgical outcomes in lesional and non-lesional epilepsy: a systematic review and meta-analysis. Epilepsy Res. 2010;89:310–8.PubMedCrossRef
147.
go back to reference Englot D, Berger M, Barbaro N, Chang E. Predictors of seizure freedom after resection of supratentorial low-grade gloiomas. A review J Neurosurg. 2011;115:240–4. Englot D, Berger M, Barbaro N, Chang E. Predictors of seizure freedom after resection of supratentorial low-grade gloiomas. A review J Neurosurg. 2011;115:240–4.
148.
go back to reference Bulacio J, Jehi L, Wong C, Gonzalez-Martinez J, Kotagal P, Nair D, et al. Long-term seizure outcome after resective surgery in patients evaluated with intracranial electrodes. Epilepsia. 2012;53:1722–30. Bulacio J, Jehi L, Wong C, Gonzalez-Martinez J, Kotagal P, Nair D, et al. Long-term seizure outcome after resective surgery in patients evaluated with intracranial electrodes. Epilepsia. 2012;53:1722–30.
149.
go back to reference Ozlen F, Gunduz A, Asan Z, Tanriverdi T, Ozkara C, Yeni N, et al. Dysembrioplastic neuroepithelial tumors and gangliogliomas: clinical results of 52 patients. Acta Neurochir. 2010;152:1661–71. Ozlen F, Gunduz A, Asan Z, Tanriverdi T, Ozkara C, Yeni N, et al. Dysembrioplastic neuroepithelial tumors and gangliogliomas: clinical results of 52 patients. Acta Neurochir. 2010;152:1661–71.
150.
go back to reference You G, Huang L, Yang P, Zhang W, Yan W, Wang Y, et al. Clinical and molecular genetic factors affecting postoperative seizure control of 183 Chinese adult patiens with low-grade gliomas. Eur J Neurol. 2012;19:298–306. You G, Huang L, Yang P, Zhang W, Yan W, Wang Y, et al. Clinical and molecular genetic factors affecting postoperative seizure control of 183 Chinese adult patiens with low-grade gliomas. Eur J Neurol. 2012;19:298–306.
151.
go back to reference Gump W, Skjei K, Karkare S. Seizure control after subtotal lesional resection. Neurosurg Focus. 2013;34:E1.PubMedCrossRef Gump W, Skjei K, Karkare S. Seizure control after subtotal lesional resection. Neurosurg Focus. 2013;34:E1.PubMedCrossRef
152.
go back to reference Englot D, Han S, Berger M, Barbaro N, Chang E. Extent of surgical resection predicts seizure freedom in low-grade temporal lobe brain tumors. Neurosurgery. 2012a;70:921–8.PubMedCrossRef Englot D, Han S, Berger M, Barbaro N, Chang E. Extent of surgical resection predicts seizure freedom in low-grade temporal lobe brain tumors. Neurosurgery. 2012a;70:921–8.PubMedCrossRef
153.
go back to reference Englot D, Berger M, Barbaro N, Chang E. Factors associated with seizure freedom in the surgical resection of glioneuronal tumors. Epilepsia. 2012b;53:51–7.PubMedCrossRef Englot D, Berger M, Barbaro N, Chang E. Factors associated with seizure freedom in the surgical resection of glioneuronal tumors. Epilepsia. 2012b;53:51–7.PubMedCrossRef
156.
go back to reference • Mittal S, Shah A, Barkmeier D, Loeb J. Systems biology of human epilepsy applied to patients with brain tumors. Epilepsia. 2013;54(Suppl 9):35–9. This paper provides an expalantion of the systems biology approach developed to study patients with tumor-related epilepsy who undergo brain surgery. PubMedCrossRef • Mittal S, Shah A, Barkmeier D, Loeb J. Systems biology of human epilepsy applied to patients with brain tumors. Epilepsia. 2013;54(Suppl 9):35–9. This paper provides an expalantion of the systems biology approach developed to study patients with tumor-related epilepsy who undergo brain surgery. PubMedCrossRef
157.
go back to reference Beaumont T, Yao B, Shah A, Kapatos G, Loeb J. Layer-specific CREB target gene induction in human neocortical epilepsy. J Neurosci. 2012;32:14389–401.PubMedPubMedCentralCrossRef Beaumont T, Yao B, Shah A, Kapatos G, Loeb J. Layer-specific CREB target gene induction in human neocortical epilepsy. J Neurosci. 2012;32:14389–401.PubMedPubMedCentralCrossRef
158.
go back to reference Lipovich L, Dachet F, Cai J, Bagla S, Balan K, Jia H, et al. Activity-dependent human brain coding/noncoding gene regulatory networks. Genetics. 2012;192:1133–48. Lipovich L, Dachet F, Cai J, Bagla S, Balan K, Jia H, et al. Activity-dependent human brain coding/noncoding gene regulatory networks. Genetics. 2012;192:1133–48.
159.
go back to reference Ruda R, Bello L, Duffau H, Soffietti R. Seizures in low-grade gliomas: natural history, pathogenesis, and outcome after treatments. Neuro-Oncology. 2012;14:iv55–64.PubMedPubMedCentralCrossRef Ruda R, Bello L, Duffau H, Soffietti R. Seizures in low-grade gliomas: natural history, pathogenesis, and outcome after treatments. Neuro-Oncology. 2012;14:iv55–64.PubMedPubMedCentralCrossRef
Metadata
Title
Brain Tumor-Related Epilepsy: a Current Review of the Etiologic Basis and Diagnostic and Treatment Approaches
Author
Jeffrey M. Politsky
Publication date
01-09-2017
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 9/2017
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-017-0777-3

Other articles of this Issue 9/2017

Current Neurology and Neuroscience Reports 9/2017 Go to the issue

Dementia (K Marder, Section Editor)

New Therapeutic Strategies for Lewy Body Dementias

Epilepsy (CW Bazil, Section Editor)

Psychogenic Non-Epileptic Seizures

Demyelinating Disorders (J Bernard and M Cameron, Section Editors)

CLIPPERS

Stroke (H Diener, Section Editor)

Unresolved Issues in Thrombectomy