You are on page 1of 7

Seizure 21 (2012) 153–159

Contents lists available at SciVerse ScienceDirect

Seizure
journal homepage: www.elsevier.com/locate/yseiz

Review

The pathogenesis of tumor-related epilepsy and its implications for clinical


treatment
Gan You a, Zhiyi Sha b, Tao Jiang a,*
a
Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
b
Department of Neurology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA

A R T I C L E I N F O A B S T R A C T

Article history: Approximately 30–50% of patients with brain tumors present with seizures as the initial symptom.
Received 18 October 2011 Seizures play a very important role in the quality of life, particularly in patients with slow-growing
Received in revised form 28 December 2011 primary brain tumors. Tumor-related seizures are often refractory to antiepileptic treatment. Despite the
Accepted 29 December 2011
importance of this subject to the fields of neurology, neurosurgery and neurooncology, the pathogenesis
of tumor-related epilepsy remains poorly understood. This review summarizes possible mechanisms
Keywords: underlying the pathogenesis of tumor-related epilepsy, including both tumoral and peri-tumoral
Brain tumor
aspects. Tumor cells themselves may create intrinsic epileptogenicity, and inadequate homeostasis in
Epilepsy
Pathogenesis
the peri-tumoral tissues may lead to seizure susceptibility. Other local changes in electrolytes, perfusion,
Seizure metabolism, and enzymes could also contribute. It is generally accepted that changes in amino acid
Treatment neurotransmission are the most important mechanism underlying tumor-related seizures, and changes
in extracellular ions also play an important role. Hypoxia, acidosis, and metabolic, immunological, and
inflammatory changes may also be involved in the occurrence of seizures. Knowledge of these
mechanisms may provide guidance in the search for new strategies for the surgical and medical
treatment of tumor-related epilepsy.
ß 2012 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.

1. Introduction the tumor itself may excrete molecules that could make the tumor
tissue epileptogenic, or it could change the peri-tumoral microen-
Epileptic seizures due to brain tumors have been described vironment and turn this into an epileptogenic zone. The other
since the 19th century. John Hughling Jackson first reported the theory is that the tumor mechanically compresses the surrounding
direct relationship between seizures and brain tumor.1 Approxi- normal tissue, which eventually becomes epileptogenic after
mately 30–50% of patients with brain tumors present with seizures suffering from ischemia and hypoxia. Both of these processes could
as the initial symptom.2,3 Seizures play a very important role in the potentially cause secondary changes, such as changes in neuro-
quality of life, particularly in patients with slow-growing primary transmitters and their receptors, metabolic changes, and inflam-
brain tumors, for whom seizure incidence reaches 80–90%.2,4 matory responses, eventually leading to epileptic seizures.
These seizures often manifest as focal seizures with secondary However, neither of these theories can fully explain the patterns
generalization and are often refractory to antiepileptic treatment. of seizure incidence. This review attempts to summarize the
A number of studies have established an association between possible mechanisms of tumor-related epilepsy, based on the
epileptic seizures at disease onset and a more favorable progno- hypotheses mentioned above. This knowledge may provide
sis.4,5 Seizure onset at a later time, however, is frequently guidance in the search for new strategies for the surgical and
associated with neurosurgical procedures or progression of the medical treatment of tumor-related epilepsy.
lesion.6,7
Despite the importance of this subject to the fields of neurology, 2. Mechanisms
neurosurgery, and neurooncology, the pathogenesis of tumor-
related epilepsy remains poorly understood. At present, there 2.1. Tumor factors
seem to be two schools of thought regarding the pathogenesis of
tumor-related epilepsy. One theory is based on the tumor origin: 2.1.1. Histology
Seizures are common in patients with low-grade tumors such
as dysembryoblastic neuroepithelial tumors (DNETs), ganglioglio-
* Corresponding author. Tel.: +86 010 67036083; fax: +86 010 67036038. mas (GGs), and oligodendrogliomas (OGs). The incidence of
E-mail address: taojiang1964@yahoo.com.cn (T. Jiang). epilepsy in high-grade brain tumors such as glioblastoma multi-

1059-1311/$ – see front matter ß 2012 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.seizure.2011.12.016
154 G. You et al. / Seizure 21 (2012) 153–159

formes (GBMs) and metastatic tumors is lower. The pathogenesis accumulation of extracellular potassium, facilitation of N-methyl-
of seizure development is likely to be different for brain tumors D-aspartate (NMDA) receptor-mediated neuronal hyperexcitabili-
with a different histology.8,9 Developmental tumors consist of ty, and eventually epileptiform activity. Another study demon-
well-differentiated cells, which are able to release neurotrans- strated22 that blockade of TGF-b receptors in vivo reduced the
mitters and other modulators that are involved in epileptogen- likelihood of epileptogenesis.
esis.10 These tumors may also be associated with structural Taken together, these results suggest that pathological disrup-
epileptogenic abnormalities of the cortex. The highly infiltrative tion of the BBB in brain tumor patients may contribute to seizure
growth of fast-growing high-grade brain tumors may damage the activity. It is difficult to define the extent to which BBB
subcortical network essential for electrical transmission,11 disturbances may lead to seizure induction. However, it is
whereas slow-growing tumors have been suggested to induce generally considered that tumors such as LGGs, which cause
partial deafferentation of cortical regions, causing denervation BBB disturbances but do not destroy the subcortical network, are
hypersensitivity10,12 and producing an epileptogenic milieu. In likely to cause seizures.
addition, gliosis and chronic inflammatory changes in the peri-
tumoral regions may lead to epileptic seizures. Brain tumors with 2.1.4. Gap junctions
the same grade but a different histology may have different Communication between adjacent glial cells can take place
seizure incidences.6 Furthermore, not all patients with similar through transmembrane proteins known as connexins (CX).
tumor localization and histology have seizures.9 This strongly Astrocytic gap junctions contain CX43, whereas oligodendrocytes
suggests that genetic factors may play a role in tumor develop- express CX32, and neurons express CX26 and CX32.23 Recently,
ment and tumor-related epilepsy. alterations in glial gap-junctional coupling have been found to be
associated with tumoral epilepsy. Aronica et al.23 demonstrated
2.1.2. Tumor location increased CX43 membrane immunoreactivity in low-grade glio-
The location of the tumor is also an important determinant of mas when compared with both high-grade lesions and control
tumor-associated epilepsy. Chang et al.6 reported that subcortical brain tissue. Furthermore, the perilesional epileptogenic cortex
location with tumor growth in deep midline structures was less surrounding low-grade lesions contained reactive astrocytes that
likely to result in seizures. Some authors13 also suggested that expressed higher levels of CX43 than control cortex.23 Interesting-
location and proximity to the cortical gray matter were also ly, high-grade gliomas exhibited aberrant intracytoplasmic locali-
important factors in the development of epilepsy in patients with zation of CX43,23 which might explain the lower risk of tumor-
glioma. In general, tumors in the frontal and temporal lobes, as related seizures among patients with this type of cancer.
well as in the limbic system, are more likely to cause seizures than
tumors in other locations.6,14 The location of tumors is closely 2.1.5. Molecular genetic changes
related to their histology. The majority of glioneuronal tumors Genetic factors inside the tumor may also play a role in the
occur in the temporal lobe. Duffau and Capelle15 have provided epileptogenic process. For example, expression of tumor-suppres-
some evidence that diffuse low-grade astrocytomas and oligoden- sor gene LGI1, which could contribute to glioma progression by
drogliomas may also have preferred locations. In contrast to increasing cell growth and migration when down-regulated,24 is
malignant gliomas, low-grade gliomas (LGGs) tend to grow in low or absent in cell lines from high-grade gliomas.25 Based on the
secondary functional areas that are close to, but rarely within, the possible association between seizure recurrence and glioma
primary eloquent parts of the brain. Our unpublished data have progression,6,7 Brodtkorb et al.26 suggested that LGI1 may be
shown that oligodendroglial tumors were more likely to be located correlated with epileptic susceptibility in patients with brain
in frontal lobe, whereas astrocytomas were more commonly found tumors. However, some other studies have not supported a tumor-
in temporal locations. This might be explained by the fact that suppressor function of LGI1,27 and further investigations are
tumor histology is to some extent associated with the lobe where needed to confirm such a role.
tumors occur during brain development. Loss of heterozygosity at Tumors have genomic and chromosomal instability, including
1p/19q is closely related to tumor histology in gliomas. In a recent DNA strand breaks and rearrangements. These alterations may be
study,16 we hypothesized that some genes on 19q might be associated with changes in gene expression, with negative effects
susceptibility genes of glioma-related seizures. It may be on the stability of DNA repair mechanisms, and this may lead to
postulated that tumor-related seizures have unique character- mutations. Under these conditions, the tumor cells might become
istics, which may share some common genetic pathways with epileptogenic. Bordey and Sontheimer28 reported that astrocytic
tumorigenesis. tumor cells might generate action potentials, which might
themselves be the source of epileptic activity. Ye and Sontheimer29
2.1.3. Blood–brain barrier disruption found that glioma cells released glutamate and caused excitotoxic
The cellular components of the blood–brain barrier (BBB) cell death of the peri-tumoral neurons. These findings suggest that
include endothelial cells, astrocytes, pericytes, neurons, and the tumor itself could be an independent origin of epileptogenesis.
junctional complexes formed by transmembrane junctional Further studies of the possible candidate susceptibility genes for
proteins including occludin, claudins, and junctional adhesion tumor-related seizures are encouraged (Figs. 1 and 2).
molecules.17 Human and animal studies have suggested that
perturbations in neurovascular integrity and breakdown of the BBB 2.2. Peri-tumoral factors
lead to neuronal hypersynchronization and epileptiform activity.
Relevant molecular changes in brain tumors that affect BBB The microenvironment in brain tumors is substantially differ-
structure and function include decreased expression of transmem- ent from that in normal brain tissue. Contemporary imaging
brane junctional proteins18 and heightened release of vascular techniques provide testimony to the remarkable differences
endothelial growth factor (VEGF).19 Diffusion of VEGF into the peri- between the peri-tumoral brain and normal tissue. Recent
tumoral brain may aggravate the edema surrounding the lesion. advances in magnetic resonance spectroscopy (MRS) have
Stewart et al.20 reported structural defects in endothelial tight cell demonstrated decreased levels of N-acetylaspartate, a marker of
junctions surrounding human gliomas. A more recent study21 neuronal viability and function, in lesional epileptogenic cortex.30
suggested that this may be mediated by transforming growth Focal abnormalities of low-frequency magnetic activity on
factor b (TGF-b) receptor stimulation, causing activity-dependent magnetoencephalography (MEG) in peri-tumoral regions have
G. You et al. / Seizure 21 (2012) 153–159 155

Fig. 1. MRI (T2-weighted images) of two patients with astrocytoma (WHO grade II), suggesting different underlying biological characteristics of tumors with similar location
and histology. (a) Image from a patient that presented with secondary generalized seizures as the initial symptom. (b) Image from a patient with long-standing headache
before admission.

Fig. 2. Immunohistochemical staining for the astrocytic marker GFAP (glial fibrillary acidic protein) in the marginal areas of samples from two different patients with diffuse
astrocytomas (WHO grade II). (a) High GFAP expression in the tumor of one patient with refractory seizures. (b) Low GFAP expression in another astrocytoma of a patient who
became seizure-free on antiepileptic drugs after an initial seizure (original magnification, 400).

been found in 13 of 20 patients with tumor-related seizures.31 inhibitory synapses and increase in excitatory synapses in peri-
Douw et al. also indicated that pathologically increased theta band tumoral pyramidal neurons.35 Dysfunctional astrocytes in peri-
connectivity was related to a higher number of epileptic seizures in tumoral regions may also contribute to epilepsy through different
brain tumor patients, suggesting that elevated theta band mechanisms.36,37
connectivity is a hallmark of tumor-related epilepsy.32 Whether
these associations reflect local metabolic imbalances, functional 2.2.2. Hypoxia, acidosis and metabolic changes
deafferentation, or profound structural alterations remains un- Hypoxia and acidosis are produced as a result of intratumoral
clear. decreased perfusion and elevated metabolism, respectively.
However, the relationships between perfusion, hypoxia, metabo-
2.2.1. Morphologic changes lism, and acidosis are complex. The growth of brain tumors
Certain morphologic changes in the peri-tumoral brain tissue, depends on the establishment of an adequate blood supply.38 On
such as persistent neurons in the white matter, inefficient neuronal one hand, tumors with insufficient blood supply often cause
migration, and changes in synaptic vesicles, are also believed to interstitial hypoxia, which subsequently contributes to acidosis.
contribute to seizure generation.8,33 It is possible, and indeed This intratumoral hypoxia and acidosis may extend to the
likely, that peri-tumoral cells have an altered or anomalous surrounding tissue. On the other hand, tumors with large size
phenotype, which is commonly seen in glioneuronal or dysplastic usually cause peri-tumoral hypoxia because of direct compression.
brain tumors. This could be related to the origin of the tumor or Both these factors could cause glial cell swelling and damage.39
could constitute a predisposing factor for seizures in people with This is of particular interest because astroglial cells control the
brain tumors. Comparison of the ultrastructure of the peri-tumoral acidity of the environmental fluid. Under these conditions, the
cortex in patients with and without epilepsy has demonstrated astrocytic cell membrane becomes prone to inward sodium
statistically significant changes in the form, size, distribution, and currents, leading to risk of epilepsy.40
number of synaptic vesicles.34 Another type of cellular change Furthermore, hypoxia causes acidosis as a consequence of both
suggested to be associated with tumor growth is a decrease in heightened metabolic requirements of the proliferating tissue and
156 G. You et al. / Seizure 21 (2012) 153–159

impaired oxidative energy metabolism. The normal response to ionotropic and metabotropic glutamate receptors.53 Glutamate
hypoxia would be glucose catabolism with lactate production, receptors can be present in greater or lesser numbers in tumors,
leading to acidosis in metabolically active tissue. With the advent depending on the degree of differentiation and transcriptional
of magnetic resonance imaging (MRI), MRS, and positron emission control in the tumor tissue.
tomography, many studies have evaluated the metabolic activity Glutamate activation of ionotropic receptors leads to a rapid
in both epileptic and neoplastic brain. Increased lactate levels41 excitatory signal based on cation influx that can cause release of
and decreased glucose metabolism were observed in the peri- calcium from intracellular stores. Ionotropic glutamate receptors
tumoral white matter. An elevated level of lactate compared to include NMDA receptors (NR1, NR2A–C subunits), a-amino-3-
healthy tissue has also been reported in tumor tissue.42 Thus, the hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors
epileptogenic changes must have a component that is transferable (GluR1–4 subunits), and kainate receptors. Aronica et al.54
to the surrounding neurons. However, it is unclear how the examined 41 cases of gangliogliomas and 16 cases of DNETs, all
metabolic abnormalities described above could be responsible for with intractable epilepsy. Both tumor types had higher NR2A and
this, unless they have an effect on neuronal morphology or NR2B expression in the tumors than in the nonepileptic cortex. In a
function. similar study, Lee et al.55 evaluated NMDA receptor expression in
13 patients with DNET and refractory epilepsy. Stronger NR1,
2.2.3. Ionic changes NR2A, and NR2B expression with increased GluR2 and GluR3
Ionic changes in the peri-tumoral zone may influence neuronal immunopositivity was found in the peri-tumoral cortex. Aronica
activity. Hossmann et al.43 described elevated sodium and calcium et al.54 also demonstrated elevated expression of kainate receptors
levels in the extracellular peri-tumoral space, which may in the reactive astrocytes of the perilesional zone.
contribute to neuronal hyperexcitability. Kraft et al.44 also Interaction of glutamate with metabotropic glutamate recep-
demonstrated that, under acidic conditions, astrocytoma cell tors (mGluR) initiates intracellular signaling through GTP-
membranes are susceptible to inward sodium currents following binding proteins, which can serve neuromodulatory functions.
sodium channel activation. The relevant family of ion channels has Abnormalities of receptor activation could result from differ-
been termed the brain sodium channel complex.45 Likewise, ences in the relative expression in peri-tumoral tissue compared
increased densities and altered gating of calcium channels have to normal brains. It has been suggested that mGluR2 expression
been reported in epileptic tissue. Potassium channel mutations was decreased in the neuronal component of GGs and DNETs,
(e.g., KCNQ) have been found in certain epileptic syndromes, and while mGluR5 was expressed at higher levels on reactive
extracellular potassium concentrations are thought to play a role in astrocytes surrounding those tumors.54 Rutecki et al.56 showed
ictogenicity due to their impact on membrane potential.46,47 that metabotropic receptors might modulate AMPA receptor
Magnesium (Mg2+) helps to stabilize neuronal excitability by activation to generate oscillatory calcium waves. This effect
blocking calcium influx through NMDA receptor channels. Avoli could be inhibited by AMPA receptor blocker, but not by NMDA
et al.48 demonstrated that decreased extracellular concentrations or g-aminobutyric acid A (GABAA) receptor antagonists. In
of Mg2+ can lead to spontaneous epileptiform discharges and addition, activation of mGluR5 can lead to phosphoinositol-
spreading depression in human cortex in vitro. In addition, based calcium signaling.57 Further investigations of how
increased levels of Fe3+ in intra- or peri-tumoral areas due to metabotropic receptor expression is altered in seizure-related
small bleedings from pathological blood vessels may also tumors will be necessary to validate the pathophysiological roles
contribute to the development of tumor-associated seizures. This of these changes.
is more likely to occur in high-grade gliomas, since experimental
Fe3+-induced peroxidative injury to neuronal plasma membranes 2.2.4.2. GABA neurotransmission. The inhibitory neurotransmitter
is related to the development of paroxysmal epileptiform GABA can inhibit neuronal firing. GABA receptor down-regulation
activity.8,49 may contribute to hyperactivity of the surrounding microenviron-
Recently, an interesting hypothesis was proposed by Sonthei- ment. GABA receptors are classified as GABAA, GABAB and GABAC
mer,50 who suggested that glioma invasion into the peri-tumoral receptors. In peri-tumoral tissue, changes in the function of these
zone is in part mediated by chloride (Cl ) channel overexpres- receptors have been well documented, and mainly involve reduced
sion, allowing cells to traverse the extracellular space through GABAergic neurotransmission.55,58 Labrakakis et al.59 investigated
rapid changes in cell shape. Similar mechanisms may be involved the responsiveness of glial tumor tissue slices to GABA, and
in glioma metastasis, to cause seeding and infiltration of the observed a depolarizing response to GABAA receptor activation in
narrow extracellular spaces in the brain.51 It remains unclear 71% of oligodendroglioma cells and 62% of astrocytoma cells. In
whether seizures represent an associated feature of ion channel contrast, no activation by GABA was found in GBM specimens. This
overexpression. Further studies are needed to confirm this indicates that the GABAA receptor is expressed at higher levels in
hypothesis. LGGs compared to GBM, and that responses are often excitatory in
tumor tissue.
2.2.4. Changes in amino acids and neurotransmitter receptors Interestingly, GABA levels appeared to be higher in human
Previous studies29,52 have demonstrated that perturbations in tumor-inflicted tissues than in control tissue.46,47 Although
the balance between excitatory and inhibitory compounds may GABAergic activity does not directly correlate with seizure
lead to glioma-related seizures. Dysfunctional astrocytes may send susceptibility, it is likely that the changes in transmitter levels
signals to neurons by releasing a number of gliotransmitters that are causal for tumor-associated epilepsies. To date, the role of
can have both excitatory and inhibitory actions. GABA and its receptors in tumor-induced epilepsy is still largely
unclear.
2.2.4.1. Glutamate neurotransmission. Recent work has demon-
strated a close link between seizure activity and high extracellular 2.2.4.3. Other amino acid transmitters. Other amino acids may also
glutamate in tumor-related epilepsy. Patients with glioma and play a role in tumoral epileptogenesis. Gliomas have been shown to
refractory epilepsy have increased concentrations of glutamate, as produce lower levels of kynurenic acid,60 which may lead to
shown by changes in perilesional immunoreactivity of glutamate disinhibition of NMDA receptors.61 Decreased levels of noradren-
decarboxylase.40,52 Glutamate is an excitatory neurotransmitter aline and serotonin, which can have intrinsic inhibitory and anti-
that acts on postsynaptic membranes by interacting with epileptogenic effects, have also been demonstrated in gliomas.58
G. You et al. / Seizure 21 (2012) 153–159 157

2.2.5. Immunological and inflammatory changes preference rather than clinical evidence. However, a meta-analysis
Several studies have reported an association between strong by Glantz et al.73 of 12 informative studies investigating the use of
immunological or inflammatory changes with decreased risks of prophylactic anticonvulsants (PHT, PB, or VPA) in patients with
glioma.62,63 Immunological factors are also likely to play a role in primary and metastatic brain tumors demonstrated a lack of
tumor-associated epilepsy, and proinflammatory cytokines and efficacy in preventing the first seizure or in decreasing the
their receptors have been suggested to be involved in the frequency of initial seizures. Temkin also reported that there was
pathogenesis of epilepsy. Cytokines, such as interleukin 4 (IL-4) no evidence that long-term treatment with PHT and CBZ could
and IL-6,62 have modulating effects on neurotoxic neurotransmit- protect against late seizures.74 The question of whether or not the
ters that are released during excitation or inflammation in the choice of a traditional AED should be solely based on their side
central nervous system. IL-1b and tumor necrosis factor a effect profile remains to be answered.
interfere with astrocyte gap junction communication. In cell VPA is thought to inhibit epileptic discharges by stabilizing
culture, release of these proinflammatory cytokines from activated neuronal membranes and enhancing GABA transmission. It can
microglia leads to a closure of CX43-mediated gap junctions in induce apoptosis, growth arrest, and cell differentiation of tumor
astrocytes.64 Since status epilepticus induces massive microglial cells through inhibition of histone deacetylase.75 A recent study
activation in mice in vivo,65 inhibition of gap junction communi- reported that VPA induced autophagy in glioma cells and this
cation in astrocytes appears to represent a very early alteration in action was independent of apoptosis.76 A study by Weller et al. also
the process of epileptogenesis. In glioma, immune-mediated indicated potential anti-tumor activity of VPA in patients with
neurochemical changes by microglia in the peri-tumoral brain GBM who required an AED during temozolomide-based chemor-
regions are well documented.66,67 Immune-mediated neuronal adiotherapy.77 The fact that both tumoral and peri-tumoral factors
damage of the peri-tumoral brain area, coupled with changes in the contribute to the pathogenesis of tumor-related epilepsy suggests
balance between stimulatory and inhibitory cytokines, may that VPA should be considered as a first line therapy in treating
contribute to the development of tumor-related epilepsy.66,68 tumor-related epilepsy.

3. Treatment implications for tumor-related epilepsy 3.3. What advantages will new AEDs bring?

Seizures caused by brain tumors often manifest as focal seizures Newer generation AEDs, such as levetiracetam (LEV), lamo-
with or without secondary generalization, and approximately one trigine (LTG), topiramate (TPX), gabapentin (GBP), and pregabalin
third of patients are refractory to antiepileptic medication (PGB), have been recommended for patients with brain tumors.
treatment. There are a number of possible reasons for the These drugs have different antiepileptic mechanisms, including
medication resistance.69 First, antiepileptic drugs (AEDs) could GABA receptor agonism, calcium channel modulation, and NMDA
be affected by the biochemical milieu of the peri-tumoral space. receptor antagonism. Vecht and van Breemen12 have reviewed
Second, significant drug–drug interactions between AEDs and treatment recommendations for patients with brain tumors, and
chemotherapeutics may affect antiepileptic effectiveness, and suggested that first-line anticonvulsants should include LEV and
exaggerate side effects by influencing the hepatic cytochrome LTG, because they lack significant drug–drug interactions with
P450 system. Third, treatment resistance may also arise from chemotherapy agents. The antiepileptic mechanisms of LTG
overexpression of multidrug resistance-related proteins (MRPs) in include Na channel inhibition, inhibition of Ca2+ currents through
tumors, which restrict the penetration of lipophilic substances into neuron-specific, high-voltage-activated (N-type) Ca2+ channels,
the brain. MRPs were found at heightened levels in brain tumor enhancement of GABA receptor activity, and inhibition of
capillary endothelial cells and astrocytes.69 pathological release of glutamate and NMDA receptor-mediated
excitatory synaptic transmission. Studies evaluating the effects of
3.1. How much resection is required? LEV have found it beneficial for both monotherapy and adjunct
therapy in tumor-related epilepsy.78,79 The synaptic vesicle protein
Based on the presumption that neurons surrounding the tumor 2A (SV2A) is considered to be the binding site for LEV.80,81 Recently,
constitute the epileptogenic zone, removing the tumor alone may Lee et al.82 demonstrated that enhancing the activity of ROMK1
not guarantee a good outcome in seizure control. However, it can channels, which stabilize the resting membrane potential, may be
also be argued that if the irritating lesion is removed, the an important mechanism of LEV in preventing seizure initiation.
microenvironment may return to normal and the surrounding LEV has been shown to be very effective and well tolerated in
neurons may cease to discharge abnormally. For most surgical patients with brain tumor-associated seizures.
series involving pediatric patients, lesionectomy alone yielded In a recent study, the gap junction inhibitor carbenoxolone has
very good results.70,71 However, studies on adult patients been evaluated in organotypic hippocampal slice cultures as a
demonstrated that gross total resection or even extended potential antiepileptic agent. It was found that it inhibited both
lesionectomy could greatly improve seizure prognosis.6,72 One spontaneous and evoked seizure-like events.83 The clinical utility
possible reason for the better results of lesionectomy in children of this strategy remains uninvestigated, but it is possible that it
may be that their seizure history is shorter with less opportunity of may be valuable in the context of low-grade gliomas accompanied
permanent secondary changes such as hippocampal sclerosis. In by elevated CX43 expression.
general, surgeons are encouraged to minimize the residual tumor
volume when possible. 4. Conclusions

3.2. Should traditional AEDs be phased out? Epileptic seizures are common in patients with brain tumors.
The underlying pathogenesis of tumor-related epilepsy is not
No randomized clinical trials have evaluated the efficacy of effectively addressed by conventional AEDs. To date, the epilepto-
traditional AEDs, such as valproic acid (VPA), carbamazepine (CBZ), genic mechanisms of tumor-related epilepsy remain largely
phenytoin (PHT) and phenobarbital (PB), in patients with brain unclear. Tumor cells may create internal epileptogenicity through
tumors. It is difficult to draw firm conclusions from available their intrinsic characteristics, and inadequate homeostasis in the
studies. Hence, the decision of which AEDs to administer to peri-tumoral tissue may cause alterations in the balance between
patients with brain tumors is based mainly on individual excitation and inhibition. Other local changes in electrolytes,
158 G. You et al. / Seizure 21 (2012) 153–159

Fig. 3. Schematic of observed mechanisms underlying the pathogenesis and clinical strategies of tumor-related epilepsy, showing the possible intra- and peri-tumoral factors
causing tumor-related seizures and how current treatments take effects. (Factors are ranked in the top left corner based on their presumed importance in epileptogenesis.)

perfusion, metabolism, and enzymes could also contribute. It is 10. Wolf HK, Roos D, Blumcke I, Pietsch T, Wiestler OD. Perilesional neurochemical
changes in focal epilepsies. Acta Neuropathol 1996;91:376–84.
generally accepted that changes in neurotransmission and 11. Norden AD, Blumenfeld H. The role of subcortical structures in human epilepsy.
extracellular ion concentrations are the most important mecha- Epilepsy Behav 2002;3:219–31.
nisms underlying tumor-related seizures. Hypoxia, acidosis, and 12. Vecht CJ, van Breemen M. Optimizing therapy of seizures in patients with brain
tumors. Neurology 2006;67:S10–3.
metabolic, immunological, and inflammatory changes also play 13. Schaller B, Ruegg SJ. Brain tumor and seizures: pathophysiology and its impli-
potential roles in the induction of seizures. cations for treatment revisited. Epilepsia 2003;44:1223–32.
This review has summarized the possible pathogenesis of 14. Engel Jr J. Seizures and epilepsy. Philadelphia: FA Davis; 1989. p. 221–39.
15. Duffau H, Capelle L. Preferential brain locations of low-grade gliomas. Cancer
tumor-associated epilepsy and has also created a framework on 2004;100:2622–6.
which novel therapeutics may be applied (Fig. 3). Further 16. Huang L, You G, Jiang T, Li G, Li S, Wang Z. Correlation between tumor-related
investigations of both intra- and peri-tumoral pathophysiology seizures and molecular genetic profile in 103 Chinese patients with low-grade
gliomas: a preliminary study. J Neurol Sci 2011;302:63–7.
may give guidance in the choice of medical therapy. Increased
17. Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD. Blood–brain barrier: struc-
understanding of the dynamic processes at the tumor–brain tural components and function under physiologic and pathologic conditions. J
interface may lead to novel concepts and treatment strategies for Neuroimmune Pharmacol 2006;1:223–36.
tumor-associated epilepsy in the future. 18. Liebner S, Fischmann A, Rascher G, Duffner F, Grote EH, Kalbacher H, et al.
Claudin-1 and claudin-5 expression and tight junction morphology are altered
in blood vessels of human glioblastoma multiforme. Acta Neuropathol
2000;100:323–31.
References 19. Chi OZ, Hunter C, Liu X, Tan T, Weiss HR. Effects of VEGF on the blood–
brain barrier disruption caused by hyperosmolarity. Pharmacology 2008;82:
1. Jackson JH. Localized convulsions from tumour of the brain. Brain 1882;5:364– 187–92.
74. 20. Stewart PA, Hayakawa K, Farrell CL, Del Maestro RF. Quantitative study of
2. van Breemen MS, Wilms EB, Vecht CJ. Epilepsy in patients with brain tumours: microvessel ultrastructure in human peri-tumoral brain tissue. Evidence for a
epidemiology, mechanisms, and management. Lancet Neurol 2007;6:421–30. blood–brain barrier defect. J Neurosurg 1987;67:697–705.
3. Lote K, Stenwig AE, Skullerud K, Hirschberg H. Prevalence and prognostic 21. Ivens S, Kaufer D, Flores LP, Bechmann I, Zumsteg D, Tomkins O, et al. TGF-beta
significance of epilepsy in patients with gliomas. Eur J Cancer 1998;34:98–102. receptor mediated albumin uptake into astrocytes is involved in neocortical
4. Danfors T, Ribom R, Berntsson SG, Smits A. Epileptic seizures and survival epileptogenesis. Brain 2007;130:535–47.
during early disease in grade 2 gliomas. Eur J Neurol 2009;16:823–31. 22. Cacheaux LP, Ivens S, David Y, Lakhter AJ, Bar-Klein G, Shapira M, et al.
5. Piepmeier J, Christopher S, Spencer D, Byrne T, Kim J, Knisel JP, et al. Variations Transcriptome profiling reveals TGF-beta signaling involvement in epilepto-
in the natural history and survival of patients with supratentorial low-grade genesis. J Neurosci 2009;29:8927–35.
astrocytomas. Neurosurgery 1996;38:872–8. 23. Aronica E, Gorter JA, Jansen GH, Leenstra S, Yankaya B, Troost D, et al. Expres-
6. Chang EF, Potts MB, Keles GE, Lamborn KR, Chang SM, Barbaro NM, et al. Seizure sion of connexin 43 and connexin 32 gap-junction proteins in epilepsy-associ-
characteristics and control following resection in 332 patients with low-grade ated brain tumors and in the perilesional epileptic cortex. Acta Neuropathol
gliomas. J Neurosurg 2008;108:227–35. 2001;101:449–59.
7. Chaichana KL, Parker SL, Olivi A, Quinones-Hinojosa A. Long-term seizure 24. Kunapuli P, Chitta KS, Cowell JK. Suppression of the cell proliferation and
outcomes in adult patients undergoing primary resection of malignant brain invasion phenotypes in glioma cells by the LGI1 gene. Oncogene 2003;22:
astrocytomas. J Neurosurg 2009;111:282–92. 3985–91.
8. Beaumont A, Whittle IR. The pathogenesis of tumour associated epilepsy. Acta 25. Besleaga R, Montesinos-Rongen M, Perez-Tur J, Siebert R, Deckert M. Expression
Neurochir (Wien) 2000;142:1–15. of the LGI1 gene product in astrocytic gliomas: downregulation with malignant
9. Rosati A, Tomassini A, Pollo B, Ambrosi C, Schwarz A, Padovani A, et al. Epilepsy progression. Virchows Arch 2003;443:561–4.
in cerebral glioma: timing of appearance and histological correlations. J Neu- 26. Brodtkorb E, Nakken KO, Steinlein OK. No evidence for a seriously increased
rooncol 2009;93:395–400. malignancy risk in LGI1-caused epilepsy. Epilepsy Res 2003;56:205–8.
G. You et al. / Seizure 21 (2012) 153–159 159

27. Piepoli Z, Jakupoglu C, Gu W, Lualdi E, Suarez-Merino B, Poliani PL, et al. 57. Coutinho V, Knopfel T. Metabotropic glutamate receptors: electrical and chem-
Expression studies in gliomas and glial cells do not support a tumor suppressor ical signaling properties. Neuroscientist 2002;8:551–61.
role for LGI1. Neuro-oncol 2006;8:96–108. 58. Haglund MM, Berger MS, Kunkel DD, Franck JE, Ghatan S, Ojemann GA. Changes
28. Bordey A, Sontheimer H. Properties of human glial cells associated with in gamma-aminobutyric acid and somatostatin in epileptic cortex associated
epileptic seizure foci. Epilepsy Res 1998;32:286–303. with low-grade gliomas. J Neurosurg 1992;77:209–16.
29. Ye ZC, Sontheimer H. Glioma cells release excitotoxic concentrations of gluta- 59. Labrakakis C, Patt S, Hartmann J, Kettenmann H. Functional GABA(A) receptors
mate. Cancer Res 1999;59:4383–91. on human glioma cells. Eur J Neurosci 1998;10:231–8.
30. Chernov MF, Kubo O, Hayashi M, Izawa M, Maruyama T, Usukura M, et al. 60. Vezzani A, Gramsbergen JB, Speciale C, Schwarcz R. Production of quinolinic
Proton MRS of the peri-tumoral brain. J Neurol Sci 2005;228:137–42. acid and kynurenic acid by human gliomas. Adv Exp Med Biol 1991;294:
31. Baayen JC, de Jongh A, Stam CJ, de Munck JC, Jonkman JJ, Trenite DG, et al. 691–5.
Localization of slow wave activity in patients with tumor-associated epilepsy. 61. Hwa GG, Avoli M. The involvement of excitatory amino acids in neocortical
Brain Topogr 2003;16:85–93. epileptogenesis: NMDA & non NMDA receptors. Exp Brain Res 1991;86:248–56.
32. Douw L, van Dellen E, de Groot M, Heimans JJ, Klein M, Stam CJ, et al. Epilepsy is 62. Brenner AV, Butler MA, Wang SS, Ruder AM, Rothman N, Schulte PA, et al.
related to theta band brain connectivity and network topology in brain tumor Single-nucleotide polymorphisms in selected cytokine genes and risk of adult
patients. BMC Neurosci 2010;11:103. glioma. Carcinogenesis 2007;28:2543–7.
33. Villemure JG, de Tribolet N. Epilepsy in patients with central nervous system 63. Wiemels JL, Wiencke JK, Kelsey KT, Moghadassi M, Rice T, Urayama KY, et al.
tumors. Curr Opin Neurol 1996;9:424–8. Allergy-related polymorphisms influence glioma status and serum IgE levels.
34. Chubinidze AI, Gobechiia ZV, Abramishvili VV, Samodurova GV, Chubinidze Cancer Epidemiol Biomarkers Prev 2007;16:1229–35.
MA. Morphological characteristics of cortical synapses in patients with epilep- 64. Meme W, Calvo CF, Froger N, Ezan P, Amigou E, Koulakoff A, et al. Proinflam-
sy. Zh Nevropatol Psikhiatr Im S S Korsakova 1985;89:23–6. matory cytokines released from microglia inhibit gap junctions in astrocytes:
35. McNamara JO. Emerging insights into the genesis of epilepsy. Nature 1999;399: potentiation by beta-amyloid. FASEB J 2006;20:494–6.
A15–22. 65. Avignone E, Ulmann L, Levavasseur F, Rassendren F, Audinat E. Status epilepti-
36. De Keyser J, Mostert JP, Koch MW. Dysfunctional astrocytes as key players in the cus induces a particular microglial activation state characterized by enhanced
pathogenesis of central nervous system disorders. J Neurol Sci 2008;267:3–16. purinergic signaling. J Neurosci 2008;28:9133–44.
37. Seiferta G, Carmignotob G, Steinhäusera C. Astrocyte dysfunction in epilepsy. 66. Chao CC, Molitor TW, Hu S. Neuroprotective role of IL-4 against activated
Brain Res Rev 2010;63:212–21. microglia. J Immunol 1993;151:1473–81.
38. Fidler IJ, Yano S, Zhang RD, Fujimaki T, Bucana CD. The seed and soil hypothesis: 67. Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the
vascularization and brain metastasis. Lancet Oncol 2002;3:53–7. molecular mechanisms. Nat Rev Neurosci 2007;8:57–69.
39. Kempski O, Staub F, Jansen M, Schodel F, Baethmann A. Glial swelling during 68. Hulkkonen J, Koskikallio E, Rainesalo S, Keranen T, Hurme M, Peltola J. The
extracellular acidosis in vitro. Stroke 1988;19:385–92. balance of inhibitory and excitatory cytokines is differently regulated in vivo
40. Schaller B. Influences of brain tumor-associated pH changes and hypoxia on and in vitro among therapy resistant epilepsy patients. Epilepsy Res
epileptogenesis. Acta Neurol Scand 2005;111:75–83. 2004;59:199–205.
41. Herholz K, HeindelW. Luyten PR, denHollander JA, Pietrzyk U, Voges J, et al. In 69. Löscher W. How to explain multidrug resistance in epilepsy? Epilepsy Curr
vivo imaging of glucose consumption and lactate concentration in human 2005;5:107–12.
gliomas. Ann Neurol 1992;31:319–27. 70. Kim SK, Wang KC, Hwang YS, Kim KJ, Cho BK. Intractable epilepsy associated
42. Martinez-Outschoorn UE, Prisco M, Ertel A, Tsirigos A, Lin Z, Pavlides S, et al. with brain tumors in children: surgical modality and outcome. Childs Nerv Syst
Ketones and lactate increase cancer cell stemness, driving recurrence, metas- 2001;17:445–52.
tasis and poor clinical outcome in breast cancer. Cell Cycle 2011;10:1271–86. 71. Giulioni M, Galassi E, Zucchelli M, Volpi L. Seizure outcome of lesionectomy in
43. Hossmann KA, Seo K, Szymas J, Wechsler W. Quantitative analysis of experi- glioneuronal tumors associated with epilepsy in children. J Neurosurg
mental peri-tumoral edema in cats. Adv Neurol 1990;52:449–58. 2005;102:288–93.
44. Kraft R, Basrai D, Benndorf K, Patt S. Serum deprivation and NGF induce and 72. Hildebrand J, Lecaille C, Perennes J, Delattre JY. Epileptic seizures during follow-
modulate voltage-gated Na(+) currents in human astrocytoma cell lines. Glia up of patients treated for primary brain tumors. Neurology 2005;65:212–5.
2001;34:59–67. 73. Glantz MJ, Cole BF, Forsyth PA, Recht LD, Wen PY, Chamberlain MC, et al.
45. Berdiev BK, Xia J, McLean LA, Markert JM, Gillespie GY, Mapstone TB, et al. Acid- Practice parameter: anticonvulsant prophylaxis in patients with newly diag-
sensing ion channels in malignant gliomas. J Biol Chem 2003;278:15023–34. nosed brain tumors. Report of the Quality Standards Subcommittee of the
46. Avoli M, Louvel J, Pumain R, Köhling R. Cellular and molecular mechanisms of American Academy of Neurology. Neurology 2000;54:1886–93.
epilepsy in the human brain. Prog Neurobiol 2005;77:166–200. 74. Temkin NR. Antiepileptogenesis and seizure prevention trials with antiepileptic
47. Badawy RA, Harvey AS, Macdonell RA. Cortical hyperexcitability and epilepto- drugs: meta-analysis of controlled trials. Epilepsia 2001;42:515–24.
genesis: understanding the mechanisms of epilepsy—part 1. J Clin Neurosci 75. Li XN, Shu Q, Su JM, Perlaky L, Blaney SM, Lau CC. Valproic acid induces growth
2009;16:355–65. arrest, apoptosis, and senescence in medulloblastomas by increasing histone
48. Avoli M, Drapeau C, Louvel J, Pumain R, Olivier A, Villemure JG. Epileptiform hyperacetylation and regulating expression of p21Cip1, CDK4, and CMYC. Mol
activity induced by low extracellular magnesium in the human cortex main- Cancer Ther 2005;4:1912–22.
tained in vitro. Ann Neurol 1991;30:589–96. 76. Fu J, Shao CJ, Chen FR, Ng HK, Chen ZP. Autophagy induced by valproic acid is
49. Singh R, Pathak DN. Lipid peroxidation and glutathione peroxidase, glutathione associated with oxidative stress in glioma cell lines. Neuro-oncol 2010;12:
reductase, superoxide dismutase, catalase and glucose-6-phosphate dehydro- 328–40.
genase activities in FeCl3 induced epileptogenic foci in the rat brain. Epilepsia 77. Weller M, Gorlia T, Cairncross JG, van den Bent MJ, Mason W, Belanger K, et al.
1990;31:15–26. Prolonged survival with valproic acid use in the EORCT/NCIC temozolomide
50. Sontheimer H. Ion channels and amino acid transporters support the growth trial for glioblastoma. Neurology 2011;77:1156–64.
and invasion of primary brain tumors. Mol Neurobiol 2004;29:61–71. 78. Dinapoli L, Maschio M, Jandolo B, Fabi A, Pace A, Sperati F, et al. Quality of life
51. Sontheimer H. An unexpected role for ion channels in brain tumor metastasis. and seizure control in patients with brain tumor-related epilepsy treated with
Exp Biol Med (Maywood) 2008;233:779–91. levetiracetam monotherapy: preliminary data of an open-label study. Neurol Sci
52. Bateman DE, Hardy JA, McDermott JR, Parker DS, Edwardson JA. Amino acid 2009;30:353–9.
transmitter levels in gliomas and their relationship to the incidence of epilepsy. 79. Lee JW, Bromfield EB, Kesari S. Antiemetic properties of the antiepileptic drug
Neurol Res 1988;10:112–4. levetiracetam. N Engl J Med 2008;359:1853.
53. Xiao MY, Gustafsson B, Niu YP. Metabotropic glutamate receptors in the 80. Gillard M, Chatelain P, Fuks B. Binding characteristics of levetiracetam to
trafficking of ionotropic glutamate and GABAA receptors at central synapses. synaptic vesicle protein 2A (SV2A) in human brain and in CHO cells expressing
Curr Neuropharmacol 2006;4:77–86. the human recombinant protein. Eur J Pharmacol 2006;536:102–8.
54. Aronica E, Yankaya B, Jansen GH, Leenstra S, van Veelen CW, Gorter JA, et al. 81. de Groot M, Aronica E, Heimans JJ, Reijneveld JC. Synaptic vesicle protein 2A
Ionotropic and metabotropic glutamate receptor protein expression in glio- predicts response to levetiracetam in patients with glioma. Neurology
neuronal tumours from patients with intractable epilepsy. Neuropathol Appl 2011;77:532–9.
Neurobiol 2001;27:223–37. 82. Lee CH, Lee CY, Tsai TS, Liou HH. PKA-mediated phosphorylation is a novel
55. Lee MC, Kang JY, Seol MB, Kim HS, Woo JY, Lee JS, et al. Clinical features and mechanism for levetiracetam, an antiepileptic drug, activating ROMK1 chan-
epileptogenesis of dysembryoplastic neuroepithelial tumor. Childs Nerv Syst nels. Biochem Pharmacol 2008;76:225–35.
2006;22:1611–8. 83. Samoilova M, Wentlandt K, Adamchik Y, Velumian AA, Carlen PL. Connexin 43
56. Rutecki PA, Sayin U, Yang Y, Hadar E. Determinants of ictal epileptiform mimetic peptides inhibit spontaneous epileptiform activity in organotypic
patterns in the hippocampal slice. Epilepsia 2002;43(Suppl. 5):179–83. hippocampal slice cultures. Exp Neurol 2008;210:762–75.

You might also like