Open Access

Seizures in brain tumors: pathogenesis, risk factors and management (Review)

  • Authors:
    • Cyrille D. Dantio
    • Deborah Oluwatosin Fasoranti
    • Chubei Teng
    • Xuejun Li
  • View Affiliations

  • Published online on: March 21, 2025     https://doi.org/10.3892/ijmm.2025.5523
  • Article Number: 82
  • Copyright: © Dantio et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Seizures in the context of brain tumors are a relatively common symptom, with higher occurrence rates observed in glioneuronal tumors and gliomas. It is a serious burden that can have a significant impact on the quality of life (QoL) of patients and influence the disease's prognosis. Brain tumor‑related epilepsy (BTRE) is a challenging entity because the pathophysiological mechanisms are not fully understood yet. Nonetheless, neuroinflammation is considered to play a pivotal role. Next to neuroinflammation, findings on the pathogenesis of BTRE have established that certain genetic mutations are involved, of which the most known would be IDH mutations in gliomas. Others discussed more thoroughly in the present review include genes such as PTEN, TP53, IGSF3, and these findings all provide fresh and fascinating insights into the pathogenesis of BTRE. Treatment for BTRE presents unique challenges, mainly related to burdens of polytherapy, debated necessity of anti‑epileptic prophylaxis, and overall impact on the QoL. In fact, there are no established anti‑seizure medications (ASMs) of choice for BTRE, nor is there any protocol to guide the use of these medications at every step of disease progression. Treatment strategies aimed at the tumor, that is surgical procedures, radio‑ and chemotherapy appear to influence seizure control. Conversely, some ASMs have also shown antitumor properties. The present review summarizes and retrospectively analyzes the literature on the pathogenesis and management of BTRE to provide an updated comprehensive understanding. Furthermore, the challenges and opportunities for developing future therapies aimed at BTRE are discussed.

Introduction

Annually, >300,000 brain tumor cases are diagnosed globally, making them the fifth most prevalent type of cancer. Meningiomas represent the most prevalent primary benign tumors of the central nervous system (CNS). Recent figures indicate that 40.8% of all CNS tumors in the USA are meningiomas, constituting 56.2% of all benign tumors, whereas gliomas account for 26.3% of all tumors and 50.9% of all malignant tumors (1). Brain metastases (BM) constitute a distinct category and are the most prevalent form of brain tumor, with an incidence ~5-fold greater than that of primary brain tumors (2). Lung cancers, breast cancers and melanomas are the most likely to metastasize to the brain (2). The incidence of seizures varies significantly among different types of brain tumors, ranging from 10% to over 80%, contingent upon the tumor type (3,4). This is a significant side effect that should not be underestimated due to its potential impact on the patients' quality of life (QoL). Numerous studies conducted throughout the years have endeavored to elucidate the pathophysiological mechanisms of brain tumor-related epilepsy (BTRE), yielding various degrees of results. It is generally acknowledged that mutations in the isocitrate dehydrogenase type 1 (IDH1) and type 2 genes contribute to the development of BTRE in gliomas at the molecular level (5,6). In general, various mechanisms, including mechanical (compression), vascular (imbalance in vascularization), chemical (neurotransmitter dysregulation), and inflammatory processes, have been identified in the pathophysiology of BTRE (7). Neurosurgeons frequently prescribe anti-seizure medications (ASMs) during the perioperative period, particularly post-operatively, despite the absence of studies demonstrating the advantages of this practice for seizure-naïve patients. Current research on BTRE has predominantly focused on gliomas and glioneuronal tumors, while other tumor forms, including meningiomas and BM, have received far less attention. The present review examines the pathophysiology of BTRE, encompassing molecular mechanisms and genetic implications. Risk factors, the correlation between epileptogenesis and tumorigenesis, and the influence of ASMs on tumor progression were also discussed. Finally, some insight was provided into anti-epileptogenesis, which aims to cure epilepsy. The progress made so far and the challenges associated with developing novel therapies are discussed; and the potential solutions that may be advantageous for epilepsy broadly and BTRE specifically are delineated.

Pathophysiology of BTRE

The role of neuroinflammation

Inflammation has always played a role in epilepsy, as evidenced by the existence of febrile seizures. This indicates a mechanism through which inflammation induces a hyper-excitable state in brain tissue. Numerous studies have sought to elucidate this mechanism, and while the findings remain partially unsatisfactory, the principal pathophysiological agents have been identified. In neuroinflammation, the microglia and astrocytes secrete pro-inflammatory cytokines (PICs) such as interleukin 1 beta (IL-1β), tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), among others. The overload of PICs in the CNS leads to the blood-brain barrier (BBB) breakdown, resulting in further recruitment of PICs from the systemic circulation (8,9). This review focuses on the role of IL-1β, TNF-α, toll-like receptor 4 (TLR4) and High-mobility group box 1 (HMGB1) in BTRE pathophysiology.

Il-1β can induce hyperexcitability via several mechanisms and possesses various receptors, with IL-1R1 being the most involved in epileptogenesis. Upon binding of Il-1β to IL-1R1, ceramide is generated through the activation of neutral sphingomyelinase (N-SMase). Ceramide stimulates phosphorylation of the NR2B subunit of N-methyl-D-aspartate receptors (NMDAR), leading to an increased influx of Ca2+ in neuron cells and, subsequently, increased excitability (8). This increase in intracellular calcium (Ca2+) may also stimulate the overproduction of nitric oxide (NO) through nitric oxide synthase. NO induces oxidative stress and cell damage, therefore leading to increased PICs' secretion (10). IL-1β may also induce epilepsy via the synaptic protein synaptophysin (SYN). Using rat models, Xiao et al (11) proposed that SYN can regulate neurotransmitter release by acting on Ca2+, and discovered that IL-1β increases SYN expression in the hippocampal neurons via the activation of the PI3K/Akt/mTOR pathway. In another study on temporal lobe epilepsy (TLE), using hippocampal tissues from human subjects, researchers demonstrated that the complex Il-1β/IL-1R1 was responsible for a decrease of up to 30% in GABA-mediated neurotransmission, with protein kinase C also contributing to this phenomenon (12).

TNF-α is secreted by microglial cells and astrocytes under physiological conditions to palliate an eventual decrease of glutamate, thus maintaining an adequate level of neuronal excitability (13). TNF-α has two receptors, TNFR1 and TNFR2, which are considered to have opposing functions in epilepsy (14). TNFR1 serves as the proconvulsive receptor, evidenced by neuroinflammation where excessive TNF-α-TNRF1 binding results in increased glutamate levels through various mechanisms, such as the upregulation of glutaminase in microglia and the upregulation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPAR) (15,16). TNFR1 is also responsible for the neurotoxicity effects of TNF-α due to its possession of a death domain, the TNFR-associated death domain (TADD), which is lacking in TNFR2. TADD facilitates the activation of caspase enzymes (caspases 8 and 10), resulting in cell death. While studies still remain ambiguous on the predominant pathway during TNF-α activation, some discovered that low doses of TNF-α triggered the TNF-α/TNFR1 pathway, while higher doses were required for anticonvulsant effects (TNF-α/TNFR2 pathway) (17,18). While TNF-α enhances the hyperactivity of AMPAR and NMDAR, it conversely induces endocytosis of GABA receptors, increasing GABA absorption and thus resulting in hyperexcitability (19-21).

Another inflammatory mediator with a key role in neuroinflammation would be TLRs. In immunology, TLRs function by binding to specific molecules known as damage-associated molecular patterns. One such molecule is the HMGB1, a DNA-binding protein. In pathological conditions, excess HMGB1 is produced by glial cells and neurons (among others), a reaction that is amplified by cytokines. HMGB1 acts by binding to molecules known as pattern recognition receptors, specifically TLR4 and the receptor for advanced glycated end-products (RAGE). It is considered that TLR4 has a more significant role in epileptogenesis than RAGE (22). Regardless of its binding to RAGE or TLR4, HMGB1 ultimately induces the release and activation of multiple transcription factors (23,24), with nuclear factor kappa B (NF-κB), being a prominent factor in both pathways, crucial in inflammatory and immune gene expression. Activated TLR4 can also enhance calcium influx in neurons through NMDAR, a mechanism that involves N-SMases (8,25). Moreover, HMGB1 is involved in the BBB breakdown, either through binding to RAGE or TLR4 (26), and studies reported decreased seizure activity upon inhibition of the HMGB1-TLR4 pathway (27). HMGB1 protein can also bind to Il-1β, thereby activating it. The latter has similar intracellular domains to TLR4s, and thus is involved in similar metabolic pathways (8). Il-1β and TLR4 can enhance the activation of pro-inflammatory genes through the stimulation of the NF-κB transcription factor. NOD-like receptor protein 3 is among the several activated genes. The latter activates caspase-1 via its inflammasome, which mediates inflammation and is responsible for the production of PICs. The synthesized PICs can then activate the Il-1β/TLR4 pathway, resulting in a cycle of sustained inflammation, and increased seizure risk (28-31).

Neurotransmitter disbalances

Alternative mechanisms can lead to an excessive release of glutamate or inadequate levels of GABA. The Xc-Cystine glutamate antiporter system in the brain imports cystine into the cell and in exchange, glutamate is released. Glioma cells not only express Xc-Cystine channels but due to mechanical compression and oxidative stress on adjacent tissues, there is overexpression of these channels on normal cells in an attempt to supply adequate cystine for glutathione synthesis (32). This would ultimately result in an excessive release of Glu in the synaptic space (Fig. 1). Furthermore, a comparison of glutamate transport between normal and malignant astrocytes revealed a significant deficiency of sodium-dependent reuptake channels, namely excitatory amino acid transporters 1 and 2 (EAAT1 and EAAT2) (33,34). Moreover, research has indicated a complex mechanism for glutamate transport from glioma or metastatic tumor cells to neurons, utilizing structures similar to synapses found in a normally functioning brain (35) (Fig. 2). In another study on gliomas, glutamate was found to directly down-regulate GABA receptors on astrocytes, resulting in a loss of GABAergic inhibition (36).

The role of gene expression in BTRE

IDH1 mutation in low-grade gliomas and secondary glioblastomas

IDH, comprising 3 isoforms, IDH1, IDH2 and IDH3, plays a crucial role in the Krebs cycle, by facilitating the conversion of isocitrate to α-ketoglutarate (α-KG). In anaerobic conditions, lactate dehydrogenase A (LDHA) catalyzes the synthesis of L-lactate from pyruvate. LDHA levels are increased in most tumor types, and tumor cells utilize the enzyme to increase glycolysis and lactate production rates, even under optimal aerobic conditions. This phenomenon is known as the Warburg effect (37). Missense mutations in the IDH1 gene in gliomas results in the production of 2-hydroxyglutarate (2-HG) from α-KG (38). 2-HG is considered to facilitate seizures by enhancing mTOR activity for metabolic functions such as the production of LDHA (high levels of LDHA were observed in IDH1-mutated tissues treated with 2-HG) (39). The proposed mechanism is that 2-HG elevates levels of ribosomal protein S6, which plays a role in mTOR signaling (39). Other mechanisms by which 2-HG may induce epilepsy remain contentious. A study indicated that 2-HG, owing to its structural resemblance to glutamate, may imitate its function by binding to NMDARs, hence enhancing ion influx into neuronal cells and increasing seizure susceptibility (6). The role of 2-HG in epileptogenesis is further substantiated by a condition known as 2-hydroxyglutaric aciduria, characterized by elevated levels of 2-HG, with seizures that exacerbate as the disease progresses being one of the primary symptoms.

Tumor suppressor genes PTEN, TP53, NF1 and glioblastoma

The aggressive and anarchic growth pattern of glioblastomas (GBM) results in genetic mutations that can be detected even intratumorally (40). Tumor suppressor genes phosphatase and tensin homolog (PTEN), tumor protein p53 (TP53) and Neurofibromin 1 (NF1) are affected by these mutations (41,42). It has been suggested that these changes promote tumorigenesis and subsequent tumor progression, while also contributing to epileptogenesis (43). In a healthy organism, PTEN downregulates the PI3K/AKT/mTOR pathway; however, in the context of GBM, a loss-of-function mutation in PTEN results in the disinhibition of this pathway, leading to excessive mTOR signaling (44). The role of PTEN in epileptogenesis was further suggested when scientists discovered that the targeted deletion of this gene generated seizures in animal models (45,46).

The NF1 gene, however, exerts a negative regulatory effect on the Ras (Ras/MAPK and Ras/PI3K) pathways, which are additional pathways involved in cell cycle activity. In GBM, a loss-of-function mutation in the NF1 gene results in the disinhibition of the Ras/MAPK pathway, which subsequently increases mTOR signaling (47). A study by Sabetghadam et al (48) showed that silencing of the NF1 gene in rats resulted in lowered seizure thresholds and increased severity.

TP53 (also known as P53) mutations in GBM are predominantly gain-of-function mutations, producing an altered P53 protein known as mut-P53. The latter stimulates the activation of receptor tyrosine kinases such as MNNG HOS transforming gene and EGFR (41), ultimately resulting in cell proliferation. The significance of P53 in epileptogenesis remains incompletely elucidated. Increased expression of P53 has been observed in rats and patients with TLE, particularly in the hippocampus (49,50). Engel et al (51) observed that following the triggering of epilepsy in experimental models, P53 levels increased significantly; this increase subsequently activates apoptotic and neuronal cell death processes, exacerbating imbalances and enhancing hyperexcitability. The study also demonstrated that the inhibition of P53 results in more severe seizures. This contrasts with the study by Burla et al (52) where P53 inhibition resulted in decreased seizures and inflammation. Nonetheless, inhibition of P53 has shown neuroprotective effects in a variety of conditions, including seizure-induced neuronal cell death (53,54). Further research is required to elucidate the role of P53 in epileptogenesis.

IGSF3 Gene mutation in glioma: The potassium hypothesis

Some studies showed that astrocytes are involved in potassium (K+) buffering following synaptic depolarization, via inwardly rectifying K+ channels (Kir 4.1) (55). Mutations or dysregulation of Kir 4.1 (also known as KCJN10) have been linked to seizures in various epileptic syndromes (56,57). IGSF3 protein is a cell surface protein involved in cellular signaling and molecular binding among other functions. A recent study by Curry et al (58) found that mutations in IGSF3 (gain-of-function mutation) contributed to neuronal hyperexcitability. Following the binding of a mutated IGSF3 to Kir 4.1, there is inhibition of the latter's ability to buffer K+, resulting in synaptic remodeling and depolarization spreading (58). Previous studies linked extracellular K+ concentration to ictogenesis (59). This represents a novel research direction on the mechanism of epileptogenesis in brain tumors, and further studies are encouraged to evaluate the IGFS3 gene as a potential therapeutic target in BTRE.

Risk factors for seizures in brain tumors

Preoperative risk factors for seizures in brain tumors include sex, tumor location and size, and the presence of peritumoral edema. Postoperative seizures are primarily associated with the extent of resection, the presence of preoperative seizures, and again, tumor size and location.

The occipital lobe exhibits the lowest propensity for epilepsy, whereas tumors located in the frontal and temporal lobes present the greatest seizure risk (60). Similarly, skull base tumors are less frequently associated with seizures compared with tumors located nearer to the cortex, such as convexity or parasagittal/parafalcine tumors.

Nonetheless, certain risk factors vary amongst different tumor types. IDH mutations in gliomas increase the incidence of seizures in younger patients, as low-grade gliomas (LGGs) are more prevalent in this demographic group. Regarding gliomas, a study revealed that high-grade gliomas associated with seizures were generally smaller, whereas low-grade gliomas were typically larger (61). This contrasts with meningiomas, where an increase in size correlates with a higher likelihood of seizures (62).

A significant component, other than location and size, associated with seizures in meningiomas is peritumoral edema. At a pathophysiological level, peritumoral edema is considered to contribute to a reduction in seizure threshold due to excess secretion of substances such as VEGF and glutamate (63). The male sex is linked to an increased risk of seizures in cases of meningiomas and brain metastasis (64,65). This is comprehensible, as the primary cancers that are most prone to spread to the brain are lung cancers and melanomas (66), and men are at an increased risk of developing lung cancers due to a greater prevalence of smoking and high-risk employment. The meningioma case is interesting as the tumor occurs more frequently in women than in men, with an approximate 3 to 1 ratio (1).

Brain metastasis exhibits a unique profile with specific risk factors exclusive to this category. Multiple metastasis heightens the risk of seizures in BM, as it increases the probability of a metastasis being located in a susceptible region. Studies have also shown that patients with melanoma or lung cancer as their primary tumor had an increased risk of developing seizures in case of brain invasion (67,68). In the case of melanomas, one rationale is that they are among the metastatic tumors most prone to induce intracranial hemorrhage and tumor bleeding, which is a risk factor for seizures in BM (68,69). Furthermore, a previous study by Urban et al (70) examined the impact of immune checkpoint inhibitors (ICIs) on epileptogenesis associated with brain metastasis. The study highlighted that new-onset status epilepticus occurred more frequently in patients receiving ICIs compared with those not undergoing this treatment (70), although it is important to note that the majority of patients in the ICIs group had melanoma as their primary malignancy, while lung cancer was the predominant pathology in the non-ICIs group. Additionally, a retrospective analysis of 348 patients with brain metastasis by Garcia et al (68) identified ICIs as a risk factor for late postoperative seizures. Overall, further research on this topic is necessary.

The extent of resection significantly influences the incidence of postoperative seizures across all tumor types. Complete resection diminishes the likelihood of postoperative seizure persistence. ASM prophylaxis has shown no impact on the risk of post-operative seizure, which may explain the rationale behind experts' recommendations against its usage in seizure-naïve patients. New-onset postoperative seizures are relatively rare and seem to be more closely associated with tumor grade, tumor recurrence/progression, and IDH1 mutation in glioma cases (71).

Seizures prognosis in brain tumors

It is estimated that 60-90% of patients with preoperative seizures due to brain tumors attain seizure freedom post-surgery, with glioneuronal tumors exhibiting the most favorable epilepsy prognosis, whereas seizures associated with glioblastomas tend to be more refractory and recurrent (4). Furthermore, seizures at the onset of disease serve as a favorable prognostic factor for long-term survival in brain tumors (72-74). Convexity tumors are more prone to be symptomatic in the early stages due to cortical irritation, facilitating earlier detection and intervention before progression to an advanced stage; total resection is also more attainable in these instances. Plus, IDH mutations are predominant in slow-growing, low-grade tumors, with a favorable prognosis compared with the more aggressive, high-grade ones. However, persistent seizures are a negative prognostic factor, as they are often indicative of tumor recurrence or progression (74).

De novo seizures are a relatively rare phenomenon after brain surgery, and research on this is notably limited. Danish researchers conducted a pivotal study revealing that the one-year incidence of de novo post-craniotomy epilepsy ranged from 3.8% in cases of congenital malformation to 27.6% in cases of cerebral abscess. The risk for intracranial tumors was 15.4%. This correlates with the 12.3% new seizure rate reported in the Englot et al (3) meta-analysis involving 4,709 meningioma patients in 2015. However, regarding the Danish study, the occurrence of de novo seizures varied among tumor types, and their incidence increased with time (75). Another study focused on meningiomas and de novo postoperative tonic-clonic seizures identified cerebral edema enlargement and hemorrhagic transformation of the edema as risk factors for the seizures (76). The role of surgery recurrence in postoperative de novo seizures remains contentious.

Current management of BTRE

Surgery and radio/chemotherapy

Surgery is a mainstay in the management of brain tumors and influences seizure freedom, if seizures are part of the disease burden. Gross total resection has shown its efficacy in seizure control compared with partial resection in LGGs, meningiomas and BM (77,78). In GBM, the proliferative characteristics of the tumor may cause the diseased tissue to extend beyond the boundaries shown on a contrast-enhanced MRI. Thus, supra-total resection has shown greater efficacy in seizure control in GBM compared with gross total resection (79). In addition, a study about the relationship between the extent of resection and its impact on seizure freedom in temporal lobe LGGs and glioneuronal tumors revealed that gross total resection combined with hippocampectomy showed a higher seizure freedom rate compared with gross total resection alone (80), potentially suggesting a new strategy for managing BTRE in that specific region of the brain. Further studies are needed.

Over the years, numerous studies have proven the efficacy of radiotherapy on seizure control in LGGs (81-83), alongside chemotherapy with temozolomide (84,85). There appears to be less of a consensus for glioblastomas, with some studies suggesting radiotherapy is beneficial, while others reported an increase in seizures post-radiation (86). The systematic review by Wu et al (87) also identified seizures as a side effect of radiotherapy in higher-grade meningiomas (grades II and III), however with a low incidence. Acute seizures are a known risk associated with brain irradiation resulting from neuronal and vascular alterations that may ultimately lead to brain edema and can occur several months post-radiotherapy (88,89). The impact of radio/chemotherapy on seizure control in patients with brain tumors remains poorly researched, maybe accounting for the aforementioned contradictory findings. Further research is encouraged to evaluate the risk-benefit ratio of initiating radiotherapy in brain tumor patients with epilepsy.

Anti-seizure medications
Classification and indications

The use of ASMs in the context of BTRE poses a unique challenge. In addition to potential adverse side effects, the physician should also consider probable interactions with other anticancer therapies. The ideal medications should consequently be those that alleviate both of these difficulties. BTRE is categorized as focal seizures, with or without secondary generalization (4). Multiple professional organizations have advised against the administration of ASMs as a prophylactic treatment in seizure-naïve patients with brain lesions and have discredited the need to use ASMs for patients undergoing brain tumor surgery in the absence of seizures (90). However, it is not consistently implemented in practice (91,92).

The International League Against Epilepsy classification of some ASMs is as follows: Levetiracetam (LEV), Carbamazepine (CBZ), Phenytoin (PHT) and Zonisamide (ZSD) are classified as level A ASMs. At level B, Valproic Acid (VPA) is classified; at level C, Gabapentin (GBP), Lamotrigine (LMG), Oxcarbazepine (OXC), Phenobarbital (PHB), Topiramate (TOP) and Vigabatrin (VGB) are classified; and at level D, Clonazepam (CPM) and Primidone (PMD) (93). LEV has shown superior efficacy and fewer side effects compared with other ASMs for BTRE, establishing it as the preferable treatment for monotherapy, in addition to its being a class A anticonvulsant (94,95). Moreover, LEV has no pharmacological interactions with other drugs and has minimal enzymatic activity via cytochrome P450, making it suitable for poly-medicated patients (96). Some commonly used ASMs, their side effects, and their role in the management of BTRE are summarized in Table I; whereas the mechanism of action of the ASMs predominantly employed in BTRE instances is demonstrated in Fig. 3. Refractory seizures occur in ~15-40% of people with epilepsy due to brain tumors or other etiologies (97,98). In this case, an adjunctive ASM is typically implemented rather than altering the overall treatment regimen. Polytherapy combines drugs with different mechanisms of action to maximize the results. The combination of LEV and VPA has shown significant results in cases of drug-resistant seizures (99). It should be highlighted that ASM polytherapy should be avoided in patients with BTRE wherever possible. It can significantly impact the QoL of these patients. Some studies have in fact shown that patients on multiple ASMs exhibit a higher prevalence of psychiatric comorbidities and more frequently compared with those on monotherapy (100,101).

Table I

Anti-epileptic drugs and their characteristics.

Table I

Anti-epileptic drugs and their characteristics.

ASMsGenerationMechanism of actionCommonly reported adverse side effectsSerious adverse side effectsAdditional notes(Ref.) showing its efficacy in BTRE monotherapy
Levetiracetam2ndBinds to synaptic protein SVA2Anxiety, fatigue, mood disorders, depressionSuicide ideation (extremely rare)Vit B6 shows efficacy in countering the psychiatric side effects (102)(94,95)
Valproic acid1stNa+ and Ca2+ channels blocking; GABA enhancementWeight gain, hair loss, hyperammonemia, transaminitis, tremor, rash, fatigueThrombocytopenia, hepatotoxicity, SJSRisk of hepatotoxicity and thrombocytopenia increased with chemotherapy agents (103,104); enzymatic induction(105)
Lacosamide3rdNa+ Channel blockingDrowsiness, headache, dizziness, fatiguePR prolongation, ataxia, SJS, rash, syncopeContraindicated in patient with 2nd or 3rd degree heart block (106)(107,108)
Lamotrigine2ndNa+ channel blockingCardiac conduction abnormalitiesBronchospasm, SJS, rash, angioedema, CNS depression blood dyscrasiasOne of the most common drugs to cause SJSNot reported, Add-on therapy only (109)
Topiramate2nd-AMPA antagonism
-Na+ channel blocking
-GABA enhancement
Dizziness, fatigue, anorexia, irritability, confusion, metabolic acidosis, weight lossAcute angle closure glaucoma, nephrolithiasis, SJS, rashPotential agent for the treatment of obesity (110)(111)
Zonisamide2ndNa+ and Ca2+ channels blockadeSomnolence, weight loss, rash, renal calculi, dizziness, fatigueNephrolithiasis, SJS, rash, glaucomaAvoid in patients with sulfa allergiesNo studies, add-on only (112)
Phenytoin1stNa+ channel blockingTransaminitis, rash, gingival hyperplasia, nausea, GI tract irritation, hirsutismSJS, rash, DRESS syndrome, hepatotoxicity,ASM most associated with skin-related adverse reactions (113,114); teratogenic, responsible for a condition called Fetal Hydantoin SyndromeNot reported
Carbamazepine1stNa+ channel blockingdizziness, ataxia, slurred speech, vertigo, leukopenia, agranulocytosis, skin rashAplastic anemia, hepatotoxicity, hyponatremia, SJS, rash, DRESS syndromeLong-term use (>10 years) may lead to decreased bone density; enzymatic inducer; may cause bone marrow suppressionNot reported
Oxcarbazepine2ndNa+ channel blockingFatigue, lightheadedness, weight gain, alopeciaHyponatremia, SJS, rashCytochrome enzyme interactions(115,116)
Eslicarbazepine3rdNa+ channel blockingHyponatremia, somnolence, nausea, fatigue, rashSJS, rash, hepatotoxicity, hyponatremia, blood dyscrasiasNone, Add-on only (117)
Perampanel3rdAMPA antagonismDizziness, vertigo, fatigue, aggressiveness, agitation, irritability, anxiety, nauseaSuicidal ideationQuickly emerging as a first-line monotherapy alternative for BTRE, more studies needed(118)
Gabapentin2ndA2δ Ca channel subunit inhibitionDizziness, fatigue, peripheral edema, weight.Not reported, add-on only (119)
Cenobamate3rdGABA enhancement, Na+ channel blockingHyperkalQT shortening, DRESSCytochrome enzymes interactionNot reported
Clobazam1stGABA-A-receptor agonismFatigue, dizziness, nausea, dry mouthSomnolence, sedationNot reported, add-on only (120)
Tiagabine2ndGABA enhancementGI track upset (diarrhea), fatigue, dizziness, anxiety, tremor, depressionCNS depression, rashSide effects commonly reportedNot reported, add-on only (121)
Brivaracetam3rdSVA2 bindingFatigue, dizziness, anxiety, depression, irritabilityHypersensitivity reactionsNot reported; Add-on only (122)

[i] DRESS, Drug reaction with eosinophilia and systemic symptoms (severe adverse drug reaction with extensive skin rash, visceral organ failure, lymphadenopathy, eosinophilia and atypical lymphocytosis); SJS, Steven-Johnson Syndrome; CNS, central nervous system; ASM, anti-seizure medication; GABA, gamma-aminobutyric acid; GI, gastrointestinal; BTRE, brain tumor-related epilepsy; AMPA, α-am ino-3-hydroxy-5-methyl-4-isoxazole-propionic acid.

PRN exhibits a noteworthy profile regarding BTRE. In the United States, it is utilized in monotherapy against focal-onset seizures, with or without generalized tonic-clonic seizures. AMPA antagonism is a relatively novel concept in anti-epilepsy healthcare, with the first ASMs employing this mechanism emerging in the 2nd generation. The majority of ASMs available are ion channel blockers or GABA enhancers, and over time, there is a possibility for drug resistance to occur. Targeting NMDA receptors has so far produced limited outcomes, as they facilitate slow and long-lasting synaptic changes, which are essential for learning and memory processes. As a result, researchers have shifted their attention to AMPA receptor blockade. The most ubiquitous AMPA receptor antagonist, PRN, a 3rd generation ASM, holds significant potential. In clinical trials, it has shown minimal adverse reactions, with dizziness being the most common (123,124). Currently, the clinical dosage of PRN ranges from 2 to 12 mg daily, with an increased likelihood of adverse side effects at higher doses, while the improvements in efficacy do not appear to increase proportionally (124). Although PRN is utilized as monotherapy for epilepsy in a few countries, its efficacy in BTRE has predominantly been demonstrated when used as an adjunctive ASM. A recent study reported promising results when using PRN monotherapy in patients with brain tumors, although the sample size was limited (118). Future studies with larger cohorts are needed.

Drug on drug interactions

Enzyme-inducing ASMs (EIASMs) include PHT, PHB, CBZ, PMD, OXC and TOP. VPA functions as an enzyme inhibitor. EIASMs can shorten the bioavailability of other ASMs in polytherapy; however, this is often not challenging, as the additional drug compensates for the diminished efficacy of the others. Yet still, physicians should bear these interactions in mind when implementing anti-seizure polytherapy, particularly with ASMs such as LMG that pose further challenges in clinical use.

The most sensible area for meticulous monitoring of these interactions is chemotherapy. A systematic review by Bénit and Vecht (125) estimated that EIASMs could increase the clearance of chemotherapeutic agents by 2 to 3-fold the normal rate. Glucocorticoids are also a mainstay in the treatment plan for patients with brain tumor. A study showed that dexamethasone (DXM) bioavailability was only 33% in neurosurgical patients on phenytoin, whereas it was 84% in those not receiving any ASMs (126). The inverse effect also occurs; DXM as an enzyme inducer can reduce PHT levels while also increasing it through a protein-binding mechanism (88). VPA is still routinely used in BTRE monotherapy and exhibits notable drug interactions, such as increasing the toxicity of nitrosoureas, cisplatin and etoposide (103). Inversely, cisplatin reduces the bioavailability of VPA, possibly affecting seizure control (127). All these challenges have resulted in the emergence of new classes of ASMs with minimal enzymatic activity, encouraging physicians to choose these over older generations. The chemotherapeutic drug temozolomide also presents an effective profile in glioma treatment due to its dearth of pharmacologic interactions with ASMs, as it is not metabolized by hepatic enzymes.

Comorbidities

Patients with brain tumors often present additional health complications that require extra precautions while using ASMs. Furthermore, the management of comorbidities may occasionally benefit from certain anti-seizure medications. Patients with brain tumors who also experience migraine headaches may benefit from VPA, ZSD and TOP (128,129). In the mentally disabled, ASM monotherapy should be prioritized whenever feasible due to the higher susceptibility of this population to the adverse effects of ASMs (130). Patients with behavioral issues should avoid LEV, Brivaracetam, PRN and TOP. VPA appears to be well tolerated in patients with psychiatric problems such as anxiety, depression, or psychosis, but LEV should be avoided in this group (130).

Concerning infectious comorbidities, anti-tuberculosis drugs Isoniazid and Rifampicin exert opposing effects on the metabolism of VPA (131). Numerous studies present conflicting results on the impact of VPA on viral load in patients with HIV (132-134). Further studies with larger cohorts are needed to evaluate potential interactions between VPA and antiretroviral drugs. In patients with HIV, LEV should be prioritized, with TOP as a second line option (130).

Seizures resulting from brain tumors during pregnancy are thankfully a rare phenomenon. PHT is a highly teratogenic ASM, responsible for the Fetal Hydantoin Syndrome; it should be considered a last-line option of treatment during pregnancy. Studies suggest that VPA is also responsible for numerous birth defects. In a large French cohort study by Blotiere et al (135), they investigated the risk of developing 23 distinct malformations due to prenatal exposure to 10 different ASMs; VPA emerged as the most teratogenic, with TOP also identified as a significant risk factor. ASMs suitable for BTRE monotherapy, such as LEV, OXC and the adjunct ASM LMG, were the least likely to cause birth defects (135). These results corroborated the findings of a previous study on the same topic (136). Furthermore, VPA would also be responsible for developmental and cognitive impairments in children born to mothers who received this ASM during pregnancy (137,138).

Antitumor effect of ASMs

Some of the mechanisms underlying epileptogenesis are also involved in tumor growth and progression. Tumor-to-neuron synapses act bidirectionally; the tumor increases neuronal excitability by upregulating AMPA receptor activity and glutamate release, while neurons release a mitogen known as neuroligin-3, which stimulates tumor growth through the activation of the PI3K-mTOR pathway (35,139,140). Further evidence of this mechanism was shown when AMPA receptor antagonist Perampanel (PRN) successfully slowed down tumor growth (140). Another growth mechanism is considered to involve NMDA receptors, as blockade of these receptors was successful in mitigating tumor growth in another study (141). This then presupposes that tumor treatment should influence epileptic activity, and vice-versa. Over the years, studies have emerged about the potential role of ASMs in improving survivability in brain tumor patients, to varying degrees of results. The majority of the studies have focused on LEV and VPA, due to their greater potential.

LEV has been shown to inhibit O6-Methylguanine-DNA Methyltransferase (MGMT), a DNA repair enzyme involved in the proliferation of cancer cells (142). A few retrospective studies concluded that LEV improved survival in patients undergoing chemotherapy with temozolomide for glioblastoma (143,144). However, a meta-analysis and a prospective randomized control trial, both featuring larger sample sizes and more curated statistical methodologies, found no evidence supporting the efficacy of LEV as an antineoplastic agent in patients with GBM (145,146). The differences in outcome across these studies suggest the potential influence of additional factors, such as the molecular profile of the GBM (MGMT methylated vs non-methylated; IDH-mutant vs. IDH-wild type). This presents a promising avenue for future research.

The role of VPA as an antineoplastic agent is less controversial than that of LEV, and it is the most extensively researched in this context, possibly due to the initial identification of potential beneficial effects of ASMs on survival in brain tumors involving this drug (147). Survival rate is increased when combining VPA with temozolomide for glioblastoma management (85,148,149). VPA can act as a histone deacetylase inhibitor (HDACI), particularly at high doses (150,151). HDACIs are a new group of anticancer agents that induce cell cycle arrest and eventually apoptosis in cancer cells. Another hypothesis is that VPA, through its enzyme inhibitory activity, increases the bioavailability of temozolomide, hence augmenting its chemotherapeutic efficacy (149).

Other ASMs have displayed antitumor effects, mostly in a preclinical setting. Salmaggi et al (152) and Lange et al (153) investigated the influence of PRN on tumor growth in vitro. Both studies concluded that PRN limits tumor growth by promoting cell apoptosis (152,153). Interestingly, Lange et al (154) discovered no advantage in survival improvement regarding the role of PRN on tumor progression in vivo (154). Brivaracetam and Lacosamide (LCM) have also demonstrated antineoplastic properties in in vitro experiments (155). Additionally, studies indicated that LCM and CBZ exhibit HDAC inhibition activity (156,157), suggesting a similar mechanism to VPA in inhibiting tumor growth; this requires further exploration. Other studies have shown the antiproliferative effects of LMG and PHT on breast cancers (158,159). Future studies could help determine if these two ASMs should be prioritized for seizures resulting from BM related to a breast tumor.

Withdrawal of ASMs

There is no consensus on the timing of discontinuation of ASMs in patients with BTRE, necessitating cooperation between the patient and their physician. Factors influencing ASM withdrawal include patient preference, cognitive or significant side effects, polypharmacy, and sedation or fatigue. Notable risk factors for postoperative seizures include a history of preoperative seizures, tumor progression, incomplete surgical resection and tumor location, particularly in the temporal, insular and frontal lobes.

In a prospective observational study, Koekkoek et al (160) proposed that the cessation of ASM medication should be evaluated based on tumor progression risk. The study revealed that 26% of patients who discontinued ASMs experienced recurrent seizures, with 58% of these individuals exhibiting tumor progression. In another study by Das et al (161), the effects of ASM discontinuation were examined in patients who underwent surgery for LGGs and meningiomas. Among the 111 patients who either had their medications withdrawn or never initiated anti-seizure therapy, only 9.9% experienced seizures. Notably, postoperative seizures occurred in 28% of patients who maintained uninterrupted ASMs treatment (161). This highlights the clinician's decision to continue ASMs in patients with risk factors for postoperative seizures.

The timing of withdrawal is also important. For non-tumor-related epilepsy, epileptologists suggest it is safe to discontinue ASMs after 1 year of seizure freedom; nevertheless, this choice varies significantly across clinicians (162,163). Research on tumor-associated epilepsy is limited. In a study by Jiang et al (164) on seizure relapse in glioma patients, 28 patients experienced relapse, with 20 of them (71.4%) relapsing within 6 months following the withdrawal of ASMs. All patients included in the study were on anti-seizure medication for at least 2 years postoperatively prior to withdrawal (164). In summary, withdrawal remains a contentious topic with no consensus; it is hoped that future studies could help guide practitioners in decision-making about discontinuation of ASMs.

New avenues for the management of BTRE

There is ongoing research focused on new targets for the development of ASMs. While the majority of this research is centered on primary epilepsy, certain mechanisms described in the physiopathology section of the present review are relevant to epilepsy due to causes other than brain tumors. This suggests that prospective new molecules resulting from these studies may still be beneficial to patients suffering from BTRE. Some new molecules, under investigation, and targeting mechanisms outlined in the pathophysiology section of this review are discussed below. Ongoing initiatives are being made to cure epilepsy by directly targeting epileptogenesis to alter the disease process or develop preventive measures.

Targeting new pathways Targeting isocitrate dehydrogenase and glycolysis

Generally, tumor cells utilize more energy than normal for their metabolism compared with normal cells. With IDH mutation, glioma cells mostly utilize glycolysis for ATP production, rather than the Krebs cycle (Warburg effect), and they upregulate GLUT1 and GLUT3 transporters expression to further augment energy production (165). 2-deoxy-D-glucose (2DG), a glycolysis inhibitor, is currently the focus of several preclinical trials for the treatment of various cancers, including GBM (166). PET imaging studies with radiolabeled 2DG have shown increased glucose utilization in epileptic brain regions during the ictal phase of seizures, correlating with increased neuronal activity (167). This suggests a possible anti-seizure effect of 2DG. Both the acute and chronic anti-seizure effects of 2DG were evidenced in a study by. Stafstrom et al (168), whilst another study found that long-term administration of the drug was pro-convulsant (169). A proposed rationale for these contradictory results was that 2DG, by blocking glucose entry into cells, could lead to seizures, as hypoglycemia-induced seizures are a well-documented phenomenon (169). Additionally, some cardiac side effects have been reported in preclinical trials involving 2DG (170), further undermining its safety profile for clinical application.

Ivosidenib is an IDH1 inhibitor approved for treating acute myeloid leukemia. Its efficacy as an ASM is currently confined to a limited number of case reports, with seizure control being a secondary objective in one instance (171,172). It is anticipated that these reports will stimulate further interest in the function of this molecule as an anti-seizure agent and result in more comprehensive and definitive research.

Targeting the agents of neuroinflammation

HMGB1 overexpression during neuroinflammation leads to downstream activation of PICs such as TLR4, and other factors such as receptor for RAGE and NF-κB. It is then stipulated that the HMGB1/TLR4/RAGE/NF-κB signaling pathway is responsible for the overexpression of P-glycoprotein (P-gp), a transporter protein associated with drug-resistant epilepsy (DRE) (173,174). This has led to various studies on HMGB1 blockage for the treatment of DRE, including the development of the monoclonal antibody anti-HMGB1 mAb, which has proved its anti-seizure potential in animal models (175-177). Anti-HMGB1 mAb has also shown its efficacy in a variety of CNS diseases, some of which may entail seizures (178).

BBB breakdown secondary to neuroinflammation positions astrocytes as well as PICs as potential new targets for the development of new ASMs. VX-765 (Belnacasan), an inhibitor of interleukin converting enzyme/caspase-1, which subsequently inhibits IL-1β and HMGB1 downstream, has shown anti-seizure efficacy (179), and possesses a favorable clinical profile (180). In other studies, the antagonism of TLR4 and IL-1R1 with Anakinra, a second-line drug used in the treatment of rheumatoid arthritis, diminished seizure activity in animal models (27,181,182), presumably via decreased expression of NMDA glutamate receptors. Anakinra has mostly been utilized in the care of febrile infection-related epilepsy syndrome in humans, yielding positive results (183-185). Most of the evidence derives from case reports; hence, studies with larger cohorts may be warranted. Elsewhere, research has shown that the activation of the ALK5/TGF-β signaling pathway in astrocytes, facilitated by albumin following BBB breakdown, promotes seizures via excitatory synaptogenesis (186). The mechanism involves the overactivity of this pathway, which inhibits K+ buffering by decreasing Kir 4.1 channel function and glutamate reuptake abilities of astrocytes (187). The ALK5/TGF-β pathway then constitutes another target for the development of novel anti-seizure medications.

Another prospect for inhibition in the fight against seizures and epilepsy is the complement receptor C5ar1. Complement activation is a critical occurrence in inflammation, and the study by Benson et al (188) found that pro-inflammatory receptor C5ar1 was upregulated in rats following induced status epilepticus. Utilizing the C5ar1 inhibitor PMX35, they effectively diminished both the frequency and severity of acute and chronic seizures (188). The proposed mechanism posits that the inhibition of C5ar1 results in a decreased release of PICs, including TNF-α and IL-1β.

The mTOR pathway inhibitors

The mammalian target of rapamycin (mTOR) plays a crucial role in several phases of the cell cycle and is primarily composed of two subunit complexes: mTORC1 and mTORC2. The complex mTORC1 facilitates the overexpression of AMPA receptors in vitro (189). mTOR inhibitors, such as rapamycin, have shown antiepileptic properties in animal models across multiple studies (190,191). Nonetheless, other studies have indicated that rapamycin exhibits inconsistency in its efficacy against seizures (192,193). The extensive range of mTOR signaling targets in the brain may explain the inconsistent findings among these studies. Everolimus, a rapamycin derivative, utilized in the treatment of some cancers, has shown efficacy in refractory seizures (194,195), and is currently used for the management of seizures in patients with tuberous sclerosis complex (196). Other mTOR inhibitors currently studied for their anti-seizure effects include the immunosuppressant mycophenolate, novel drugs PQR620 and PQR530, as well as natural compounds curcumin and resveratrol (197-201). PQR620 and PQR530 showed enhanced efficacy compared with rapamycin and everolimus for the management of seizures due to their expedited penetration of brain tissues (199).

Improving experimental models and approach to development of new ASMs

Researchers mostly rely on animal models to elucidate the pathogenesis of epilepsy and develop new drugs. Given that BTRE seizures are classified as focal with secondary generalization, the animal model most suitable for research is the kindling epilepsy model (202). However, the predominant models for ASM screenings are the pentylenetetrazol and maximum electroshock models (acute models). Experiments with these two models were unable to demonstrate the antiepileptic characteristics of LEV, characteristics that were ultimately identified using the kindling model (202). Due to financial constraints, the kindling model is not widely utilized; yet, to develop new therapeutic agents targeted specifically at BTRE, it is imperative to devise new, more economical models that function similarly to the kindling model.

All ASMs attain their objective by diminishing neuronal excitability in the brain, and given that this activity is crucial for normal cerebral functions, most ASMs are associated with a plethora of adverse side effects. Side effects may result in non-compliance with treatment, incurring significant costs and serving as a primary factor for breakthrough of seizures (203,204). The prolonged administration of these drugs necessitates careful consideration of adverse effects. For development of new ASMs, the efficacy is consequently at risk of being jeopardized by safety concerns. The development of agents that specifically target cell populations or circuits may enhance efficacy and improve tolerance (205). A previous study concentrated on this strategy, yielding promising results (206). Furthermore, researchers have long endeavored to develop therapies that target epileptogenesis, which, in the context of BTRE, could facilitate the establishment of preventive care and/or disease-modifying therapies (DMTs). The objective is to integrate different drugs with different mechanisms of action (some may not necessarily be ASMs), to simultaneously target multiple pathways involved in epileptogenesis, and to delineate pertinent circuits (207). The Search Tool for Interacting Chemicals is an extensive database that encompasses known and predicted interactions between chemical molecules and proteins across numerous species (208), and is utilized to determine optimal pairings. Studies using this methodology yielded combinations demonstrating promising outcomes, including LEV/TOP, LEV/TOP/GBP and LEV/Atorvastatin/Ceftriaxone (209,210). Notably, none of these combos succeeded in retaining the neuroprotective effect associated with the individual administration of some of the drugs (209,210). Nevertheless, further research would significantly aid in identifying new combinations.

Discussion and conclusion

BTRE remains a crucial public health concern, with its pathophysiological mechanisms not yet fully elucidated. Neuroinflammation significantly contributes, as do genetic mutations. Mechanical compression of tumors can induce stress that results in the release of PICs from glial cells. These PICs not only alter the electrical activity of the brain through changes in ion channel traffic and receptor expression modulation, but they can also trigger the BBB's rupture to amplify inflammation. The risk of seizure development varies with the type of brain tumor, with prevalent risk factors encompassing tumor location, size, histopathology and level of resection.

Currently, no cure for epilepsy exists, and available treatments are solely symptomatic, aimed at alleviating seizures. LEV, a modulator of neurotransmitter release through synaptic vesicle 2A binding, is currently the recommended drug for BTRE. Other agents are rapidly emerging as monotherapy alternatives, with PRN receiving special attention. The minimal drug-drug interactions, selectivity for AMPARs, and antitumor properties represent an appealing profile. Numerous anti-seizure drugs are presently undergoing various stages of development, some with new mechanisms of action and others aimed at restructuring already existing ASMs (204).

In recent years, research aimed at discovering a cure for epilepsy has achieved minimal yet significant progress. Anti-epileptogenesis has recently been a focal center of discussions on epilepsy therapy. Focused on prevention and disease modification, the achievement of anti-epileptogenesis by drug combinations would signal a significant scientific breakthrough. Especially for BTRE, the prospects are intriguing: Preventive ASM therapy in a meningioma patient with large peritumoral edema, a patient with IDH1 mutated glioma, or a melanoma patient with multiple brain metastasis. However, research on preventive anti-seizure therapy is impeded by prolonged experiment durations due to the necessity of monitoring the onset of epilepsy (or its absence, indicating successful prevention), which may last for numerous years (207,211). Consequently, most endeavors in this research area are directed towards disease modification, with the aspiration of finding a cure. DMT may also mitigate the adverse side effects associated with ASMs, whose risk escalates with prolonged usage.

New therapies require evaluation, necessitating the use of animal models. Efforts have been made to create new models, and the pursuit of developing more cost-effective and efficient alternatives aimed at BTRE may be vital for this patient population. Organizations such as the Epilepsy Therapy Screening Program (ETSP), a division of the National Institute of Neurological Disorders and Stroke in the USA, have played a significant role in this field. The ETSP allows researchers globally to evaluate new compounds through its diverse array of animal models tailored for various circumstances (212,213). Animal models developed for epilepsy prevention and disease modification research have recently been incorporated into the ETSP catalog (204).

Finally, epileptogenesis and tumorigenesis are closely interconnected processes, wherein seizures induce neuronal cell death via excitotoxicity, subsequently leading to the replacement of dead neurons with tumor cells for tumor proliferation. It is hoped that future studies will facilitate the development of a new therapeutic approach that simultaneously slows down tumor progression and prevents seizures.

Availability of data and materials

Not applicable.

Authors' contributions

CDD chose the research subject, researched the literature for relevant articles and wrote the drafts. DOF performed language and grammar editing. CT and XL revised the manuscript drafts and restructured the content, and provided access to tools used to generate the figures in the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Use of artificial intelligence tools

During the preparation of this work, Quillbot artificial intelligence tool was used to improve the readability and language of the manuscript or to generate images, and subsequently, the authors revised and edited the content produced by the artificial intelligence tools as necessary, taking full responsibility for the ultimate content of the present manuscript.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82270825).

References

1 

Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the united states in 2016-2020. Neuro Oncol. 25:IV1–IV99. 2023.PubMed/NCBI

2 

Bertolini F, Spallanzani A, Fontana A, Depenni R and Luppi G: Brain metastases: An overview. CNS Oncol. 4:37–46. 2015.PubMed/NCBI

3 

Englot DJ, Magill ST, Han SJ, Chang EF, Berger MS and McDermott MW: Seizures in supratentorial meningioma: A systematic review and meta-analysis. J Neurosurg. 124:15522015.PubMed/NCBI

4 

Englot DJ, Chang EF and Vecht CJ: Epilepsy and brain tumors. Handb Clin Neurol. 134:267–285. 2016.PubMed/NCBI

5 

Easwaran TP, Lancki N, Henriquez M, Vortmeyer AO, Barbaro NM, Scholtens DM, Ahmed AU and Dey M: Molecular classification of gliomas is associated with seizure control: A retrospective analysis. Neuromolecular. 23:315–326. 2021.

6 

Chen H, Judkins J, Thomas C, Wu M, Khoury L, Benjamin CG, Pacione D, Golfinos JG, Kumthekar P and Ghamsari F: Mutant IDH1 and seizures in patients with glioma. Neurology. 88:1805–1813. 2017.PubMed/NCBI

7 

Seidel S, Wehner T, Miller D, Wellmer J, Schlegel U and Grönheit W: Brain tumor related epilepsy: Pathophysiological approaches and rational management of antiseizure medication. Neurol Res Pract. 4:452022.PubMed/NCBI

8 

Soltani Khaboushan A, Yazdanpanah N and Rezaei N: Neuroinflammation and proinflammatory cytokines in epileptogenesis. Mol Neurobiol. 59:1724–1743. 2022.PubMed/NCBI

9 

Rana A and Musto AE: The role of inflammation in the development of epilepsy. J Neuroinflammation. 15:1442018.PubMed/NCBI

10 

Zhu X, Dong J, Han B, Huang R, Zhang A, Xia Z, Chang H, Chao J and Yao H: Neuronal nitric oxide synthase contributes to PTZ kindling epilepsy-induced hippocampal endoplasmic reticulum stress and oxidative damage. Front Cell Neurosci. 11:3772017.PubMed/NCBI

11 

Xiao Z, Peng J, Wu L, Arafat A and Yin F: The effect of IL-1β on synaptophysin expression and electrophysiology of hippocampal neurons through the PI3K/Akt/mTOR signaling pathway in a rat model of mesial temporal lobe epilepsy. Neurol Res. 39:640–648. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Roseti C, van Vliet EA, Cifelli P, Ruffolo G, Baayen JC, Di Castro MA, Bertollini C, Limatola C, Aronica E, Vezzani A and Palma E: GABAA currents are decreased by IL-1β in epileptogenic tissue of patients with temporal lobe epilepsy: Implications for ictogenesis. Neurobiol Dis. 82:311–320. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Stellwagen D and Malenka RC: Synaptic scaling mediated by glial TNF-α. Nature. 440:1054–1059. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Balosso S, Ravizza T, Perego C, Peschon J, Campbell IL, De Simoni MG and Vezzani A: Tumor necrosis factor-α inhibits seizures in mice via p75 receptors. Ann Neurol. 57:804–812. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Galic MA, Riazi K and Pittman QJ: Cytokines and brain excitability. Front Neuroendocrinol. 33:116–125. 2011. View Article : Google Scholar

16 

Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, Sonobe Y, Mizuno T and Suzumura A: Tumor necrosis factor-α induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner. J Biol Chem. 281:21362–21368. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Yuhas Y, Weizman A and Ashkenazi S: Bidirectional concentration-dependent effects of tumor necrosis factor alpha in Shigella dysenteriae-related seizures. Infect Immun. 71:2288–2291. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Grell M, Wajant H, Zimmermann G and Scheurich P: The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci USA. 95:570–575. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Stellwagen D, Beattie EC, Seo JY and Malenka RC: Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-α. J Neurosci. 25:3219–3228. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Fu CY, He XY, Li XF, Zhang X, Huang ZW, Li J, Chen M and Duan CZ: Nefiracetam attenuates Pro-inflammatory cytokines and GABA transporter in specific brain regions of rats with Post-ischemic seizures. Cell Physiol Biochem. 37:2023–2031. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Su J, Yin J, Qin W, Sha S, Xu J and Jiang C: Role for Pro-inflammatory cytokines in regulating expression of GABA transporter type 1 and 3 in specific brain regions of kainic Acid-induced status epilepticus. Neurochem Res. 40:621–627. 2015.PubMed/NCBI

22 

Iori V, Maroso M, Rizzi M, Iyer AM, Vertemara R, Carli M, Agresti A, Antonelli A, Bianchi ME, Aronica E, et al: Receptor for advanced glycation endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures. Neurobiol Dis. 58:102–114. 2013.PubMed/NCBI

23 

Xie J, Méndez JD, Méndez-Valenzuela V and Aguilar-Hernández MM: Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal. 25:2185–2197. 2013.PubMed/NCBI

24 

Chang ZL: Important aspects of Toll-like receptors, ligands and their signaling pathways. Inflamm Res. 59:791–808. 2010.PubMed/NCBI

25 

Balosso S, Liu J, Bianchi ME and Vezzani A: Disulfide-containing high mobility group Box-1 promotes N-Methyl-d-Aspartate receptor function and excitotoxicity by activating Toll-like receptor 4-Dependent signaling in hippocampal neurons. Antioxid Redox Signal. 21:1726–1740. 2014.

26 

Festoff BW, Sajja RK, van Dreden P and Cucullo L: HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J Neuroinflammation. 13:1942016.PubMed/NCBI

27 

Maroso M, Balosso S, Ravizza T, Liu J, Aronica E, Iyer AM, Rossetti C, Molteni M, Casalgrandi M, Manfredi AA, et al: Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat Med. 16:413–419. 2010.PubMed/NCBI

28 

Ulusoy C, Vanlı-Yavuz EN, Şanlı E, Timirci-Kahraman Ö, Yılmaz V, Bebek N, Küçükali Cİ, Baykan B and Tüzün E: Peripheral blood expression levels of inflammasome complex components in two different focal epilepsy syndromes. J Neuroimmunol. 347:5773432020.PubMed/NCBI

29 

Vezzani A, Balosso S and Ravizza T: Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol. 15:459–472. 2019.PubMed/NCBI

30 

Kigerl KA, de Rivero Vaccari JP, Dietrich WD, Popovich PG and Keane RW: Pattern recognition receptors and central nervous system repair. Exp Neurol. 258:5–16. 2014.PubMed/NCBI

31 

Vezzani A and Baram TZ: New Roles for interleukin-1 beta in the mechanisms of epilepsy. Epilepsy Curr. 7:45–50. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Kim JY, Kanai Y, Chairoungdua A, Cha SH, Matsuo H, Kim DK, Inatomi J, Sawa H, Ida Y and Endou H: Human cystine/glutamate transporter: cDNA cloning and upregulation by oxidative stress in glioma cells. Biochim Biophys Acta. 1512:335–344. 2001. View Article : Google Scholar : PubMed/NCBI

33 

de Groot J and Sontheimer H: Glutamate and the biology of gliomas. Glia. 59:1181–1189. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Savaskan NE, Fan Z, Broggini T, Buchfelder M and Eyüpoglu IY: Neurodegeneration in the brain tumor microenvironment: Glutamate in the limelight. Curr Neuropharmacol. 13:258–265. 2015. View Article : Google Scholar :

35 

Venkatesh HS, Morishita W, Geraghty AC, Silverbush D, Gillespie SM, Arzt M, Tam LT, Espenel C, Ponnuswami A, Ni L, et al: Electrical and synaptic integration of glioma into neural circuits. Nature. 573:539–545. 2019. View Article : Google Scholar : PubMed/NCBI

36 

De Groot M, Reijneveld JC, Aronica E and Heimans JJ: Epilepsy in patients with a brain tumour: Focal epilepsy requires focused treatment. Brain. 135:1002–1016. 2012. View Article : Google Scholar

37 

Kim JW and Dang CV: Cancer's molecular sweet tooth and the Warburg effect. Cancer Res. 66:8927–8930. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, et al: Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 462:739–744. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Mortazavi A, Fayed I, Bachani M, Dowdy T, Jahanipour J, Khan A, Owotade J, Walbridge S, Inati SK, Steiner J, et al: IDH-mutated gliomas promote epileptogenesis through d-2-hydroxyglutarate-dependent mTOR hyperactivation. Neuro Oncol. 24:1423–1435. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Szopa W, Burley TA, Kramer-Marek G and Kaspera W: Diagnostic and therapeutic biomarkers in glioblastoma: Current status and future perspectives. Biomed Res Int. 2017:80135752017. View Article : Google Scholar : PubMed/NCBI

41 

Zhang Y, Dube C, Gibert M Jr, Cruickshanks N, Wang B, Coughlan M, Yang Y, Setiady I, Deveau C, Saoud K, et al: The p53 pathway in glioblastoma. Cancers (Basel). 10:2972018. View Article : Google Scholar : PubMed/NCBI

42 

Smith JS, Tachibana I, Passe SM, Huntley BK, Borell TJ, Iturria N, O'Fallon JR, Schaefer PL, Scheithauer BW, James CD, et al: PTEN mutation, EGFR Amplification, and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J Natl Cancer Inst. 93:1246–1256. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Hu S, Kao HY, Yang T and Wang Y: Early and Bi-hemispheric seizure onset in a rat glioblastoma Multiforme model. Neurosci Lett. 766:1363512022. View Article : Google Scholar

44 

Venkatesan S, Lamfers MLM, Dirven CMF and Leenstra S: Genetic biomarkers of drug response for small-molecule therapeutics targeting the RTK/Ras/PI3K, p53 or Rb pathway in glioblastoma. CNS Oncol. 5:77–790. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Williams MR, De-Spenza T, Li M, Gulledge AT and Luikart BW: Hyperactivity of newborn pten Knock-out neurons results from increased excitatory synaptic drive. J Neurosci. 35:943–953. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Yonan JM, Chen KD, Baram TZ and Steward O: PTEN deletion in the adult dentate gyrus induces epilepsy. Neurobiol Dis. 203:1067362024. View Article : Google Scholar : PubMed/NCBI

47 

Hills KE, Kostarelos K and Wykes RC: Converging mechanisms of epileptogenesis and their insight in glioblastoma. Front Mol Neurosci. 15:9031152022. View Article : Google Scholar : PubMed/NCBI

48 

Sabetghadam A, Wu C, Liu J, Zhang L and Reid AY: Increased epileptogenicity in a mouse model of neurofibromatosis type 1. Exp Neurol. 331:1133732020. View Article : Google Scholar : PubMed/NCBI

49 

Engel T, Murphy BM, Schindler CK and Henshall DC: Elevated p53 and lower MDM2 expression in hippocampus from patients with intractable temporal lobe epilepsy. Epilepsy Res. 77:151–156. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Morrison RS, Wenzel HJ, Kinoshita Y, Robbins CA, Donehower LA and Schwartzkroin PA: Loss of the p53 tumor suppressor gene protects neurons from kainate-induced cell death. J Neurosci. 16:1337–1345. 1996. View Article : Google Scholar : PubMed/NCBI

51 

Engel T, Murphy BM, Hatazaki S, Jimenez-Mateos EM, Concannon CG, Woods I, Prehn JH and Henshall DC: Reduced hippocampal damage and epileptic seizures after status epilepticus in mice lacking proapoptotic Puma. FASEB J. 24:853–861. 2010. View Article : Google Scholar

52 

Burla R, La Torre M, Zanetti G, Bastianelli A, Merigliano C, Del Giudice S, Vercelli A, Di Cunto F, Boido M, Vernì F and Saggio I: P53-sensitive epileptic behavior and inflammation in Ft1 hypomorphic mice. Front Genet. 9:5812018. View Article : Google Scholar : PubMed/NCBI

53 

Culmsee C, Zhu X, Yu QS, Chan SL, Camandola S, Guo Z, Greig NH and Mattson MP: A synthetic inhibitor of p53 protects neurons against death induced by ischemic and excitotoxic insults, and amyloid β-peptide. J Neurochem. 77:220–228. 2001.PubMed/NCBI

54 

Djebali M, Lerner-Natoli M, Pascale M, Baille V, Bockaert J and Rondouin G: Molecular events involved in neuronal death induced in the mouse hippocampus by in-vivo injection of kainic acid. Brain Res Mol Brain Res. 93:190–198. 2001. View Article : Google Scholar

55 

Butt AM and Kalsi A: Inwardly rectifying potassium channels (Kir) in central nervous system glia: A special role for Kir4.1 in glial functions. J Cell Mol Med. 10:33–44. 2007. View Article : Google Scholar

56 

Nadella RK, Chellappa A, Subramaniam AG, More RP, Shetty S, Prakash S, Ratna N, Vandana VP, Purushottam M, Saini J, et al: Identification and functional characterization of two novel mutations in KCNJ10 and PI4KB in SeSAME syndrome without electrolyte imbalance. Hum Genomics. 13:532019. View Article : Google Scholar : PubMed/NCBI

57 

Reichold M, Zdebik AA, Lieberer E, Rapedius M, Schmidt K, Bandulik S, Sterner C, Tegtmeier I, Penton D, Baukrowitz T, et al: KCNJ10 gene mutations causing EAST syndrome (epilepsy, ataxia, sensorineural deafness, and tubulopathy) disrupt channel function. Proc Natl Acad Sci USA. 107:14490–14495. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Curry RN, Aiba I, Meyer J, Lozzi B, Ko Y, McDonald MF, Rosenbaum A, Cervantes A, Huang-Hobbs E, Cocito C, et al: Glioma epileptiform activity and progression are driven by IGSF3-mediated potassium dysregulation. Neuron. 111:682–695.e9. 2023. View Article : Google Scholar : PubMed/NCBI

59 

de Curtis M, Uva L, Gnatkovsky V and Librizzi L: Potassium dynamics and seizures: Why is potassium ictogenic? Epilepsy Res. 143:50–59. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Audrey C, Lim KS, Ahmad Zaki R, Narayanan V, Fong SL and Tan CT: From location to manifestation: A systematic review and meta-analysis of seizure prevalence in different brain tumor sites. Brain Disord. 14:1001462024. View Article : Google Scholar

61 

Lee JW, Wen PY, Hurwitz S, Black P, Kesari S, Drappatz J, Golby AJ, Wells WM III, Warfield SK, Kikinis R and Bromfield EB: Morphological characteristics of brain tumors causing seizures. Arch Neurol. 67:336–342. 2010.PubMed/NCBI

62 

Le VT, Nguyen AM, Pham TA and Nguyen PL: Tumor-related epilepsy and post-surgical outcomes: Tertiary hospital experience in Vietnam. Sci Rep. 13:108592023.PubMed/NCBI

63 

Elbadry Ahmed R, Tang H, Asemota A, Huang L, Boling W and Bannout F: Meningioma related Epilepsy-Pathophysiology, Pre/postoperative seizures predicators and treatment. Front Oncol. 12:9059762022.

64 

Harward SC, Rolston JD and Englot DJ: Seizures in meningioma. Handb Clin Neurol. 170:187–200. 2020.PubMed/NCBI

65 

Asano K, Hasegawa S, Matsuzaka M and Ohkuma H: Brain tumor-related epilepsy and risk factors for metastatic brain tumors: Analysis of 601 consecutive cases providing real-world data. J Neurosurg. 136:76–87. 2021.PubMed/NCBI

66 

Sankey EW, Tsvankin V, Grabowski MM, Nayar G, Batich KA, Risman A, Champion CD, Salama AKS, Goodwin CR and Fecci PE: Operative and peri-operative considerations in the management of brain metastasis. Cancer Med. 8:6809–6831. 2019.PubMed/NCBI

67 

Rudà R, Mo F and Pellerino A: Epilepsy in brain metastasis: An emerging entity. Curr Treat Options Neurol. 22:62020.PubMed/NCBI

68 

Garcia JH, Morshed RA, Chung J, Millares Chavez MA, Sudhakar V, Saggi S, Avalos LN, Gallagher A, Young JS, Daras M, et al: Factors associated with preoperative and postoperative seizures in patients undergoing resection of brain metastases. J Neurosurg. 138:19–26. 2023.

69 

Wolpert F, Lareida A, Terziev R, Grossenbacher B, Neidert MC, Roth P, Poryazova R, Imbach LL, Le Rhun E, Weller M, et al: Risk factors for the development of epilepsy in patients with brain metastases. Neuro Oncol. 22:718–728. 2020.

70 

Urban H, Willems LM, Ronellenfitsch MW, Rosenow F, Steinbach JP and Strzelczyk A: Increased occurrence of status epilepticus in patients with brain metastases and checkpoint inhibition. Oncoimmunology. 9:18515172020.PubMed/NCBI

71 

Li L, Li G, Fang S, Zhang K, Huang R, Wang Y, Zhang C, Li Y, Zhang W, Zhang Z, et al: New-Onset postoperative seizures in patients with diffuse gliomas: A risk assessment analysis. Front Neurol. 12:6825352021. View Article : Google Scholar : PubMed/NCBI

72 

Mastall M, Wolpert F, Gramatzki D, Imbach L, Becker D, Schmick A, Hertler C, Roth P, Weller M and Wirsching HG: Survival of brain tumour patients with epilepsy. Brain. 144:3322–3327. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Ge H, Di G, Yan Z, Liu D, Liu Y, Song K, Yang K, Hu X, Jiang Z, Hu X, et al: Does epilepsy always indicate worse outcomes? A longitudinal follow-up analysis of 485 glioma patients. World J Surg Oncol. 20:2972022. View Article : Google Scholar : PubMed/NCBI

74 

Vecht CJ, Kerkhof M and Duran-Pena A: Seizure prognosis in brain tumors: New insights and Evidence-based management. Oncologist. 19:751–759. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Giraldi L, Hansen JV, Wohlfahrt J, Fugleholm K, Melbye M and Munch TN: Postoperative de novo epilepsy after craniotomy: A nationwide register-based cohort study. J Neurol Neurosurg Psychiatry. 93:436–444. 2022. View Article : Google Scholar

76 

Abzalova DI, Sinkin MV, Yakovlev AA, Prirodov AV and Guekht AB: Risk factors for the development of de novo generalized tonic-clonic epileptic seizures in patients with supratentorial meningiomas after neurosurgical treatment. Neurosci Behav Physiol. 54:404–409. 2024.In Russian. View Article : Google Scholar

77 

Englot DJ, Berger MS, Barbaro NM and Chang EF: Predictors of seizure freedom after resection of supratentorial low-grade gliomas: A review. J Neurosurg. 115:240–244. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Schneider M, Güresir Á, Borger V, Hamed M, Rácz A, Vatter H, Güresir E and Schuss P: Preoperative tumor-associated epilepsy in patients with supratentorial meningioma: Factors influencing seizure outcome after meningioma surgery. J Neurosurg. 133:1655–1661. 2020. View Article : Google Scholar

79 

Jackson C, Choi J, Khalafallah AM, Price C, Bettegowda C, Lim M, Gallia G, Weingart J, Brem H and Mukherjee D: A systematic review and meta-analysis of supratotal versus gross total resection for glioblastoma. J Neurooncol. 148:419–431. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Englot DJ, Han SJ, Berger MS, Barbaro NM and Chang EF: Extent of surgical resection predicts seizure freedom in low-grade temporal lobe brain tumors. Neurosurgery. 70:921–927. 2012. View Article : Google Scholar

81 

Koekkoek JAF, Kerkhof M, Dirven L, Heimans JJ, Reijneveld JC and Taphoorn MJB: Seizure outcome after radiotherapy and chemotherapy in low-grade glioma patients: A systematic review. Neuro Oncol. 17:924–934. 2015.PubMed/NCBI

82 

Rudá R, Magliola U, Bertero L, Trevisan E, Bosa C, Mantovani C, Ricardi U, Castiglione A, Monagheddu C and Soffietti R: Seizure control following radiotherapy in patients with diffuse gliomas: A retrospective study. Neuro Oncol. 15:1739–1749. 2013.PubMed/NCBI

83 

Van Den Bent MJ, Afra D, De Witte O, Ben Hassel M, Schraub S, Hoang-Xuan K, Malmström PO, Collette L, Piérart M, Mirimanoff R, et al: Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: The EORTC 22845 randomised trial. Lancet. 366:985–990. 2005.PubMed/NCBI

84 

Brada M, Viviers L, Abson C, Hines F, Britton J, Ashley S, Sardell S, Traish D, Gonsalves A, Wilkins P and Westbury C: Phase II study of primary temozolomide chemotherapy in patients with WHO grade II gliomas. Ann Oncol. 14:1715–1721. 2003.PubMed/NCBI

85 

Sherman JH, Moldovan K, Yeoh HK, Starke RM, Pouratian N, Shaffrey ME and Schiff D: Impact of temozolomide chemotherapy on seizure frequency in patients with low-grade gliomas: Clinical article. J Neurosurg. 114:1617–1621. 2011.PubMed/NCBI

86 

Rades D, Witteler J, Trillenberg P, Olbrich D, Schild SE, Tvilsted S and Kjaer TW: Increasing seizure activity during radiation treatment for High-grade Gliomas-final results of a prospective interventional study. In Vivo. 36:2308–2313. 2022.PubMed/NCBI

87 

Wu A, Jin MC, Meola A, Wong HN and Chang SD: Efficacy and toxicity of particle radiotherapy in WHO grade II and grade III meningiomas: A systematic review. Neurosurg Focus. 46:E122019.PubMed/NCBI

88 

Grewal J, Grewal HK and Forman AD: Seizures and epilepsy in cancer: Etiologies, Evaluation, and Management. Curr Oncol Rep. 10:63–71. 2008.PubMed/NCBI

89 

Smart DD: Radiation toxicity in the central nervous system: Mechanisms and strategies for injury reduction. Semin Radiat Oncol. 27:332–339. 2017.PubMed/NCBI

90 

Walbert T, Harrison RA, Schiff D, Avila EK, Chen M, Kandula P, Lee JW, Le Rhun E, Stevens GHJ, Vogelbaum MA, et al: SNO and EANO practice guideline update: Anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Neuro Oncol. 23:1835–1844. 2021.PubMed/NCBI

91 

Van Der Meer PB, Dirven L, Van Den Bent MJ, Preusser M, Taphoorn MJB, Rudá R and Koekkoek JAF: Prescription preferences of antiepileptic drugs in brain tumor patients: An international survey among EANO members. Neurooncol Pract. 9:105–113. 2022.PubMed/NCBI

92 

Dewan MC, Thompson RC, Kalkanis SN, Barker FG and Hadjipanayis CG: Prophylactic antiepileptic drug administration following brain tumor resection: Results of a recent AANS/CNS section on tumors survey. J Neurosurg. 126:1772–1778. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Glauser T, Ben-Menachem E, Bourgeois B, Cnaan A, Guerreiro C, Kälviäinen R, Mattson R, French JA, Perucca E and Tomson T; ILAE Subcommission on AED Guidelines: Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 54:551–563. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Maschio M, Dinapoli L, Sperati F, Pace A, Fabi A, Vidiri A and Muti P: Levetiracetam monotherapy in patients with brain tumor-related epilepsy: Seizure control, safety, and quality of life. J Neurooncol. 104:205–214. 2011. View Article : Google Scholar

95 

Bähr O, Hermisson M, Rona S, Rieger J, Nussbaum S, Körtvelyessy P, Franz K, Tatagiba M, Seifert V, Weller M and Steinbach JP: Intravenous and oral levetiracetam in patients with a suspected primary brain tumor and symptomatic seizures undergoing neurosurgery: The HELLO trial. Acta Neurochir (Wien). 154:229–235. 2012. View Article : Google Scholar

96 

LaPenna P and Tormoehlen LM: The pharmacology and toxicology of Third-generation anticonvulsant drugs. J Med Toxicol. 13:329–342. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Jo J, Nevel K, Sutyla R, Smolkin M, Lopes MB and Schiff D: Predictors of early, recurrent, and intractable seizures in low-grade glioma. Neurooncol Pract. 8:40–47. 2020.

98 

Singh SP, Agarwal S and Faulkner M: Refractory status epilepticus. Ann Indian Acad Neurol. 17(Suppl 1): S32–S36. 2014. View Article : Google Scholar : PubMed/NCBI

99 

Van Breemen MSM, Rijsman RM, Taphoorn MJB, Walchenbach R, Zwinkels H and Vecht CJ: Efficacy of anti-epileptic drugs in patients with gliomas and seizures. J Neurol. 256:1519–1526. 2009. View Article : Google Scholar : PubMed/NCBI

100 

Yadav J, Singh P, Dabla S and Gupta R: Psychiatric comorbidity and quality of life in patients with epilepsy on anti-epileptic monotherapy and polytherapy. Tzu Chi Med J. 34:226–231. 2021. View Article : Google Scholar

101 

Khalid B, Waqar Z, Khan S, Ali I, Afzal N, Irfan A, Malik W, Muhammad Adil M, Saddiqa A, Khalil M and Munawar Z: Psychiatric implications of anti-seizure medications in epileptic population. BMC Neurol. 24:1662024. View Article : Google Scholar : PubMed/NCBI

102 

Besag FMC, Vasey MJ and Sen A: Current evidence for adjunct pyridoxine (vitamin B6) for the treatment of behavioral adverse effects associated with levetiracetam: A systematic review. Epilepsy Behav. 140:1090652023. View Article : Google Scholar : PubMed/NCBI

103 

Bourg V, Lebrun C, Chichmanian RM, Thomas P and Frenay M: Nitroso-urea-cisplatin-based chemotherapy associated with valproate: Increase of haematologic toxicity. Ann Oncol. 12:217–220. 2001. View Article : Google Scholar : PubMed/NCBI

104 

Simó M, Velasco R, Graus F, Verger E, Gil M, Pineda E, Blasco J and Bruna J: Impact of antiepileptic drugs on thrombocytopenia in glioblastoma patients treated with standard chemoradiotherapy. J Neurooncol. 108:451–458. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Kerkhof M, Dielemans JCM, Van Breemen MS, Zwinkels H, Walchenbach R, Taphoorn MJ and Vecht CJ: Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro Oncol. 15:961–967. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Sánchez-Villalobos JM, Aledo-Serrano Á, Villegas-Martínez I, Shaikh MF and Alcaraz M: Epilepsy treatment in neuro-oncology: A rationale for drug choice in common clinical scenarios. Front Pharmacol. 13:9912442022. View Article : Google Scholar : PubMed/NCBI

107 

Mo F, Meletti S, Belcastro V, Quadri S, Napolitano M, Bello L, Dainese F, Scarpelli M, Florindo I, Mascia A, et al: Lacosamide in monotherapy in BTRE (brain tumor-related epilepsy): Results from an Italian multicenter retrospective study. J Neurooncol. 157:551–559. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Villanueva V, Saiz-Diaz R, Toledo M, Piera A, Mauri JA, Rodriguez-Uranga JJ, López-González FJ, Gómez-Ibáñez A, Garcés M, González de la Aleja J, et al: NEOPLASM study: Real-life use of lacosamide in patients with brain tumor-related epilepsy. Epilepsy Behav. 65:25–32. 2016. View Article : Google Scholar : PubMed/NCBI

109 

van Opijnen MP, van der Meer PB, Dirven L, Fiocco M, Kouwenhoven MCM, van den Bent MJ, Taphoorn MJB and Koekkoek JAF: The effectiveness of antiepileptic drug treatment in glioma patients: Lamotrigine versus lacosamide. J Neurooncol. 154:73–81. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Wilding J, Van Gaal L, Rissanen A, Vercruysse F and Fitchet M: A randomized double-blind placebo-controlled study of the long-term efficacy and safety of topiramate in the treatment of obese subjects. Int J Obes Relat Metab Disord. 28:1399–1410. 2004.2004. View Article : Google Scholar : PubMed/NCBI

111 

Liu YT, Chen GT, Huang YC, Ho JT, Lee CC, Tsai CC and Chang CN: Effectiveness of dose-escalated topiramate monotherapy and add-on therapy in neurosurgery-related epilepsy: A prospective study. Medicine (Baltimore). 99:e237712020.PubMed/NCBI

112 

Maschio M, Dinapoli L, Zarabla A, Maialetti A, Giannarelli D, Fabi A, Vidiri A and Cantelmi T: Zonisamide in brain tumor-related epilepsy: An observational pilot study. Clin Neuropharmacol. 40:113–119. 2017.PubMed/NCBI

113 

De A, Rajagopalan M, Sarda A, Das S and Biswas P: Drug reaction with eosinophilia and systemic symptoms: An update and review of recent literature. Indian J Dermatol. 63:30–40. 2018.PubMed/NCBI

114 

Patocka J, Wu Q, Nepovimova E and Kuca K: Phenytoin-An anti-seizure drug: Overview of its chemistry, pharmacology and toxicology. Food Chem Toxicol. 142:1113932020.

115 

Mauro AM, Bomprezzi C, Morresi S, Provinciali L, Formica F, Iacoangeli M and Scerrati M: Prevention of early postoperative seizures in patients with primary brain tumors: Preliminary experience with oxcarbazepine. J Neurooncol. 81:279–285. 2007.

116 

Maschio M, Dinapoli L, Sperati F, Fabi A, Pace A, Vidiri A and Muti P: Oxcarbazepine monotherapy in patients with brain tumor-related epilepsy: Open-label pilot study for assessing the efficacy, tolerability and impact on quality of life. J Neurooncol. 106:651–656. 2012.

117 

Zoccarato M, Basile AM, Padovan M, Caccese M, Zagonel V and Lombardi G: Eslicarbazepine in patients with brain tumor-related epilepsy: A single-center experience. Int J Neurosci. 131:879–884. 2021.

118 

Hino U, Tamura R, Kosugi K, Ezaki T, Karatsu K, Yamamoto K, Tomioka A and Toda M: Optimizing perampanel monotherapy for surgically resected brain tumors. Mol Clin Oncol. 20:422024.PubMed/NCBI

119 

Perry JR and Sawka C: Add-on gabapentin for refractory seizures in patients with brain tumours. Can J Neurol Sci. 23:128–131. 1996.PubMed/NCBI

120 

Brahmbhatt N, Stupp R, Bushara O, Bachman E, Schuele SU and Templer JW: Efficacy of clobazam as add-on therapy in brain tumor-related epilepsy. J Neurooncol. 151:287–293. 2021.PubMed/NCBI

121 

Striano S, Striano P, Boccella P, Nocerino C and Bilo L: Tiagabine in glial tumors. Epilepsy Res. 49:81–85. 2002.PubMed/NCBI

122 

Maschio M, Maialetti A, Mocellini C, Domina E, Pauletto G, Costa C, Mascia A, Romoli M and Giannarelli D: Effect of brivaracetam on efficacy and tolerability in patients with brain Tumor-related epilepsy: A retrospective multicenter study. Front Neurol. 11:8132020.PubMed/NCBI

123 

Gao L, Lu Q, Wang Z, Yue W, Wang G, Shao X, Guo Y, Yi Y, Hong Z, Jiang Y, et al: Efficacy and safety of perampanel as early add-on therapy in Chinese patients with focal-onset seizures: A multicenter, open-label, single-arm study. Front Neurol. 14:12360462023.PubMed/NCBI

124 

Lavu A, Aboulatta L, Abou-Setta AM, Aloud B, Askin N, Rabbani R, Shouman W, Zarychanski R and Eltonsy S: Efficacy and safety of perampanel in epilepsy: A systematic review and meta-analysis of randomised controlled trials. Seizure. 102:54–60. 2022.PubMed/NCBI

125 

Bénit CP and Vecht CJ: Seizures and cancer: Drug interactions of anticonvulsants with chemotherapeutic agents, tyrosine kinase inhibitors and glucocorticoids. Neurooncol Pract. 3:245–260. 2016.PubMed/NCBI

126 

Chalk JB, Ridgeway K, Brophy T, Yelland JDN and Eadie MJ: Phenytoin impairs the bioavailability of dexamethasone in neurological and neurosurgical patients. J Neurol Neurosurg Psychiatry. 47:1087–1090. 1984.PubMed/NCBI

127 

Ikeda H, Murakami T, Takano M, Usui T and Kihira K: Pharmacokinetic interaction on valproic acid and recurrence of epileptic seizures during chemotherapy in an epileptic patient. Br J Clin Pharmacol. 59:593–597. 2005.PubMed/NCBI

128 

Bagnato F and Good J: The use of antiepileptics in migraine prophylaxis. Headache. 56:603–615. 2016.PubMed/NCBI

129 

Linde M, Mulleners WM, Chronicle EP and Mccrory DC: Antiepileptics other than gabapentin, pregabalin, topiramate, and valproate for the prophylaxis of episodic migraine in adults. Cochrane Database Syst Rev. 2013:CD0106082013.PubMed/NCBI

130 

Ruiz-Giménez J, Sánchez-Álvarez JC, Cañadillas-Hidalgo F and Serrano-Castro PJ: Antiepileptic treatment in patients with epilepsy and other comorbidities. Seizure. 19:375–382. 2010.PubMed/NCBI

131 

Patsalos PN and Perucca E: Clinically important drug interactions in epilepsy: Interactions between antiepileptic drugs and other drugs. Lancet Neurol. 2:473–481. 2003. View Article : Google Scholar : PubMed/NCBI

132 

Routy JP, Tremblay CL, Angel JB, Trottier B, Rouleau D, Baril JG, Harris M, Trottier S, Singer J, Chomont N, et al: Valproic acid in association with highly active antiretroviral therapy for reducing systemic HIV-1 reservoirs: Results from a multicentre randomized clinical study. HIV Med. 13:291–296. 2012. View Article : Google Scholar : PubMed/NCBI

133 

Lehrman G, Hogue IB, Palmer S, Jennings C, Spina CA, Wiegand A, Landay AL, Coombs RW, Richman DD, Mellors JW, et al: Depletion of latent HIV-1 infection in vivo: A proof-of-concept study. Lancet. 366:5492005. View Article : Google Scholar : PubMed/NCBI

134 

Sagot-Lerolle N, Lamine A, Chaix ML, Boufassa F, Aboulker JP, Costagliola D, Goujard C, Pallier C, Delfraissy JF and Lambotte O; ANRS EP39 study: Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS. 22:1125–1129. 2008. View Article : Google Scholar : PubMed/NCBI

135 

Blotière PO, Raguideau F, Weill A, Elefant E, Perthus I, Goulet V, Rouget F, Zureik M, Coste J and Dray-Spira R: Risks of 23 specific malformations associated with prenatal exposure to 10 antiepileptic drugs. Neurology. 93:e167–e180. 2019. View Article : Google Scholar : PubMed/NCBI

136 

Jentink J, Loane MA, Dolk H, Barisic I, Garne E, Morris JK and de Jong-van den Berg LT; EUROCAT Antiepileptic Study Working Group: Valproic acid monotherapy in pregnancy and major congenital malformations. N Engl J Med. 362:2185–2193. 2010. View Article : Google Scholar : PubMed/NCBI

137 

Meador KJ, Baker GA, Browning N, Cohen MJ, Bromley RL, Clayton-Smith J, Kalayjian LA, Kanner A, Liporace JD, Pennell PB, et al: Fetal antiepileptic drug exposure and cognitive outcomes at age 6 years (NEAD study): A prospective observational study. Lancet Neurol. 12:244–252. 2013. View Article : Google Scholar : PubMed/NCBI

138 

Christensen J, Grnøborg TK, Srøensen MJ, Schendel D, Parner ET, Pedersen LH and Vestergaard M: Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 309:1696–1703. 2013. View Article : Google Scholar : PubMed/NCBI

139 

Venkatesh HS, Johung TB, Caretti V, Noll A, Tang Y, Nagaraja S, Gibson EM, Mount CW, Polepalli J, Mitra SS, et al: Neuronal activity promotes glioma growth through Neuroligin-3 secretion. Cell. 161:803–816. 2015. View Article : Google Scholar : PubMed/NCBI

140 

Venkataramani V, Tanev DI, Strahle C, Studier-Fischer A, Fankhauser L, Kessler T, Körber C, Kardorff M, Ratliff M, Xie R, et al: Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature. 573:532–538. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Stepulak A, Sifringer M, Rzeski W, Endesfelder S, Gratopp A, Pohl EE, Bittigau P, Felderhoff-Mueser U, Kaindl AM, Bührer C, et al: NMDA antagonist inhibits the extracellular signal-regulated kinase pathway and suppresses cancer growth. Proc Natl Acad Sci USA. 102:15605–15610. 2005. View Article : Google Scholar : PubMed/NCBI

142 

Bobustuc GC, Baker CH, Limaye A, Jenkins WD, Pearl G, Avgeropoulos NG and Konduri SD: Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro Oncol. 12:917–927. 2010. View Article : Google Scholar : PubMed/NCBI

143 

Roh TH, Moon JH, Park HH, Kim EH, Hong CK, Kim SH, Kang SG and Chang JH: Association between survival and levetiracetam use in glioblastoma patients treated with temozolomide chemoradiotherapy. Sci Rep. 10:107832020. View Article : Google Scholar : PubMed/NCBI

144 

Ryu JY, Min KL and Chang MJ: Effect of anti-epileptic drugs on the survival of patients with glioblastoma multiforme: A retrospective, single-center study. PLoS One. 14:e02255992019. View Article : Google Scholar : PubMed/NCBI

145 

Chen JS, Clarke R, Haddad AF, Wang EJ, Lacroix M, Sarkar IN, Zand R, Chen ES and Toms SA: The effect of levetiracetam treatment on survival in patients with glioblastoma: A systematic review and meta-analysis. J Neurooncol. 156:257–267. 2022. View Article : Google Scholar : PubMed/NCBI

146 

Happold C, Gorlia T, Chinot O, Gilbert MR, Nabors LB, Wick W, Pugh SL, Hegi M, Cloughesy T, Roth P, 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. 34:731–739. 2016. View Article : Google Scholar : PubMed/NCBI

147 

Oberndorfer S, Piribauer M, Marosi C, Lahrmann H, Hitzenberger P and Grisold W: P450 enzyme inducing and non-enzyme inducing antiepileptics in glioblastoma patients treated with standard chemotherapy. J Neurooncol. 72:255–260. 2005. View Article : Google Scholar : PubMed/NCBI

148 

Krauze A, Megan M, Theresa CZ, Peter M, Shih JH, Tofilon PJ, Rowe L, Gilbert M and Camphausen K: The addition of Valproic acid to concurrent radiation therapy and temozolomide improves patient outcome: A Correlative analysis of RTOG 0525, SEER and a Phase II NCI trial. Cancer Stud Ther. 5:310382020.

149 

Weller M, Gorlia T, Cairncross JG, van den Bent MJ, Mason W, Belanger K, Brandes AA, Bogdahn U, Macdonald DR, Forsyth P, et al: Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma. Neurology. 77:1156–1164. 2011. View Article : Google Scholar : PubMed/NCBI

150 

Michaelis M, Doerr HW and Jr JC: Valproic acid as Anti-cancer drug. Curr Pharm Des. 13:3378–3393. 2007. View Article : Google Scholar : PubMed/NCBI

151 

Göttlicher M, Minucci S, Zhu P, Krämer OH, Schimpf A, Giavara S, Sleeman JP, Lo Coco F, Nervi C, Pelicci PG and Heinzel T: Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J. 20:6969–6978. 2001. View Article : Google Scholar : PubMed/NCBI

152 

Salmaggi A, Corno C, Maschio M, Donzelli S, D'urso A, Perego P and Ciusani E: Synergistic effect of perampanel and temozolomide in human glioma cell lines. J Pers Med. 11:3902021. View Article : Google Scholar : PubMed/NCBI

153 

Lange F, Weßlau K, Porath K, Hörnschemeyer MF, Bergner C, Krause BJ, Mullins CS, Linnebacher M, Köhling R and Kirschstein T: AMPA receptor antagonist perampanel affects glioblastoma cell growth and glutamate release in vitro. PLoS One. 14:e02116442019. View Article : Google Scholar : PubMed/NCBI

154 

Lange F, Hartung J, Liebelt C, Boisserée J, Resch T, Porath K, Hörnschemeyer MF, Reichart G, Sellmann T, Neubert V, et al: Perampanel Add-on to standard radiochemotherapy in vivo promotes neuroprotection in a rodent F98 glioma Model. Front Neurosci. 14:5982662020. View Article : Google Scholar : PubMed/NCBI

155 

Rizzo A, Donzelli S, Girgenti V, Sacconi A, Vasco C, Salmaggi A, Blandino G, Maschio M and Ciusani E: In vitro antineoplastic effects of brivaracetam and lacosamide on human glioma cells. J Exp Clin Cancer Res. 36:762017. View Article : Google Scholar : PubMed/NCBI

156 

Bang SR, Ambavade SD, Jagdale PG, Adkar PP, Waghmare AB and Ambavade PD: Lacosamide reduces HDAC levels in the brain and improves memory: Potential for treatment of Alzheimer's disease. Pharmacol Biochem Behav. 134:65–69. 2015. View Article : Google Scholar : PubMed/NCBI

157 

Beutler AS, Li S, Nicol R and Walsh MJ: Carbamazepine is an inhibitor of histone deacetylases. Life Sci. 76:3107–3115. 2005. View Article : Google Scholar : PubMed/NCBI

158 

Nelson M, Yang M, Dowle AA, Thomas JR and Brackenbury WJ: The sodium channel-blocking antiepileptic drug phenytoin inhibits breast tumour growth and metastasis. Mol Cancer. 14:132015. View Article : Google Scholar : PubMed/NCBI

159 

Pellegrino M, Rizza P, Nigro A, Ceraldi R, Ricci E, Perrotta I, Aquila S, Lanzino M, Andò S, Morelli C and Sisci D: FoxO3a mediates the inhibitory effects of the antiepileptic drug lamotrigine on breast cancer growth. Mol Cancer Res. 16:923–934. 2018. View Article : Google Scholar : PubMed/NCBI

160 

Koekkoek JAF, Kerkhof M, Dirven L, Heimans JJ, Postma TJ, Vos MJ, Bromberg JE, van den Bent MJ, Reijneveld JC and Taphoorn MJ: Withdrawal of antiepileptic drugs in glioma patients after long-term seizure freedom: Design of a prospective observational study. BMC Neurol. 14:1572014. View Article : Google Scholar : PubMed/NCBI

161 

Das RR, Artsy E, Hurwitz S, Wen PY, Black P, Golby A, Dworetzky B and Lee JW: Outcomes after discontinuation of antiepileptic drugs after surgery in patients with low grade brain tumors and meningiomas. J Neurooncol. 107:565–570. 2012. View Article : Google Scholar : PubMed/NCBI

162 

Tellez-Zenteno JF, Hernandez-Ronquillo L and Moien-Afshari F: Discontinuation of antiepileptic drugs after successful surgery: Who and when? Epileptic Disord. 14:363–370. 2012. View Article : Google Scholar : PubMed/NCBI

163 

Berg AT, Langfitt JT, Spencer SS and Vickrey BG: Stopping antiepileptic drugs after epilepsy surgery: A survey of U.S. epilepsy center neurologists. Epilepsy Behav. 10:219–222. 2007. View Article : Google Scholar : PubMed/NCBI

164 

Jiang H, Deng G, Liu B, Cheng J, Li Y, Tan Y, Wang J and Chen Q: Analysis of the short-term outcomes and risk factors of seizure relapse in patients with gliomas after antiepileptic drugs withdrawal. J Clin Neurosci. 82:20–25. 2020. View Article : Google Scholar : PubMed/NCBI

165 

Han W, Shi J, Cao J, Dong B and Guan W: Emerging roles and therapeutic interventions of aerobic glycolysis in glioma. Onco Targets Ther. 13:6937–6955. 2020. View Article : Google Scholar : PubMed/NCBI

166 

Singh R, Gupta V, Kumar A and Singh K: 2-Deoxy-D-Glucose: A novel pharmacological agent for killing hypoxic tumor cells, oxygen dependence-lowering in covid-19, and other pharmacological activities. Adv Pharmacol Pharm Sci. 2023:99933862023.PubMed/NCBI

167 

Siclari F, Prior JO and Rossetti AO: Ictal cerebral positron emission tomography (PET) in focal status epilepticus. Epilepsy Res. 105:356–361. 2013. View Article : Google Scholar : PubMed/NCBI

168 

Stafstrom CE, Roopra A and Sutula TP: Seizure suppression via glycolysis inhibition with 2-deoxy-D-glucose (2DG). Epilepsia. 49:97–100. 2008. View Article : Google Scholar : PubMed/NCBI

169 

Gasior M, Yankura J, Hartman AL, French A and Rogawski MA: Anticonvulsant and proconvulsant actions of 2-deoxy-d-glucose. Epilepsia. 51:1385–1394. 2010. View Article : Google Scholar : PubMed/NCBI

170 

Minor RK, Smith DL Jr, Sossong AM, Kaushik S, Poosala S, Spangler EL, Roth GS, Lane M, Allison DB, de Cabo R, et al: Chronic ingestion of 2-deoxy-D-glucose induces cardiac vacuolization and increases mortality in rats. Toxicol Appl Pharmacol. 243:332–339. 2010. View Article : Google Scholar :

171 

Tejera D, Kushnirsky M, Gultekin SH, Lu M, Steelman L and De La Fuente MI: Ivosidenib, an IDH1 inhibitor, in a patient with recurrent, IDH1-mutant glioblastoma: A case report from a Phase I study. CNS Oncol. 9:CNS622020. View Article : Google Scholar : PubMed/NCBI

172 

Vo AH, Ambady P and Spencer D: The IDH1 inhibitor ivosidenib improved seizures in a patient with drug-resistant epilepsy from IDH1 mutant oligodendroglioma. Epilepsy Behav Rep. 18:1005262022. View Article : Google Scholar : PubMed/NCBI

173 

Xie Y, Yu N, Chen Y, Zhang K, Ma HY and Di Q: HMGB1 regulates P-glycoprotein expression in status epilepticus rat brains via the RAGE/NF-κB signaling pathway. Mol Med Rep. 16:1691–1700. 2017. View Article : Google Scholar : PubMed/NCBI

174 

Chen Y, Huang XJ, Yu N, Xie Y, Zhang K, Wen F, Liu H and Di Q: HMGB1 Contributes to the Expression of P-Glycoprotein in mouse epileptic brain through Toll-like receptor 4 and receptor for advanced glycation end products. PLoS One. 10:e01409182015. View Article : Google Scholar : PubMed/NCBI

175 

Fu L, Liu K, Wake H, Teshigawara K, Yoshino T, Takahashi H, Mori S and Nishibori M: Therapeutic effects of anti-HMGB1 monoclonal antibody on pilocarpine-induced status epilepticus in mice. Sci Rep. 7:11792017. View Article : Google Scholar : PubMed/NCBI

176 

Ravizza T, Terrone G, Salamone A, Frigerio F, Balosso S, Antoine DJ and Vezzani A: High Mobility Group Box 1 is a novel pathogenic factor and a mechanistic biomarker for epilepsy. Brain Behav Immun. 72:14–21. 2018. View Article : Google Scholar

177 

Paudel YN, Othman I and Shaikh MF: Anti-high mobility group Box-1 monoclonal antibody attenuates Seizure-induced cognitive decline by suppressing neuroinflammation in an adult zebrafish model. Front Pharmacol. 11:6130092021. View Article : Google Scholar : PubMed/NCBI

178 

Nishibori M, Mori S and Takahashi HK: Anti-HMGB1 monoclonal antibody therapy for a wide range of CNS and PNS diseases. J Pharmacol Sci. 140:94–101. 2019. View Article : Google Scholar : PubMed/NCBI

179 

Maroso M, Balosso S, Ravizza T, Iori V, Wright CI, French J and Vezzani A: Interleukin-1β biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutics. 8:304–315. 2011. View Article : Google Scholar : PubMed/NCBI

180 

Kaur H, Kumar B and Medhi B: Antiepileptic drugs in development pipeline: A recent update. eNeurologicalSci. 4:42–51. 2016. View Article : Google Scholar : PubMed/NCBI

181 

Bertani I, Iori V, Trusel M, Maroso M, Foray C, Mantovani S, Tonini R, Vezzani A and Chiesa R: Inhibition of IL-1β signaling normalizes NMDA-Dependent neurotransmission and reduces seizure susceptibility in a mouse model of Creutzfeldt-Jakob disease. J Neurosci. 37:10278–10289. 2017.PubMed/NCBI

182 

Semple BD, O'Brien TJ, Gimlin K, Wright DK, Kim SE, Casillas-Espinosa PM, Webster KM, Petrou S and Noble-Haeusslein LJ: Interleukin-1 receptor in seizure susceptibility after traumatic injury to the pediatric brain. J Neurosci. 37:7864–7877. 2017.PubMed/NCBI

183 

Dilena R, Mauri E, Aronica E, Bernasconi P, Bana C, Cappelletti C, Carrabba G, Ferrero S, Giorda R, Guez S, et al: Therapeutic effect of Anakinra in the relapsing chronic phase of febrile infection-related epilepsy syndrome. Epilepsia Open. 4:344–350. 2019.PubMed/NCBI

184 

Kenney-Jung DL, Vezzani A, Kahoud RJ, LaFrance-Corey RG, Ho ML, Muskardin TW, Wirrell EC, Howe CL and Payne ET: Febrile infection-related epilepsy syndrome treated with anakinra. Ann Neurol. 80:939–945. 2016.PubMed/NCBI

185 

Dilena R, Mauri E, Aronica E, Bernasconi P, Bana C, Cappelletti C, Carrabba G, Ferrero S, Giorda R, Guez S, et al: Therapeutic effect of Anakinra in the relapsing chronic phase of febrile infection-related epilepsy syndrome. Epilepsia Open. 4:344–350. 2019.PubMed/NCBI

186 

Weissberg I, Wood L, Kamintsky L, Vazquez O, Milikovsky DZ, Alexander A, Oppenheim H, Ardizzone C, Becker A, Frigerio F, et al: Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction. Neurobiol Dis. 78:115–125. 2015.PubMed/NCBI

187 

Sanz P and Garcia-Gimeno MA: Reactive glia inflammatory signaling pathways and epilepsy. Int J Mol Sci. 21:40962020.PubMed/NCBI

188 

Benson MJ, Thomas NK, Talwar S, Hodson MP, Lynch JW, Woodruff TM and Borges K: A novel anticonvulsant mechanism via inhibition of complement receptor C5ar1 in murine epilepsy models. Neurobiol Dis. 76:87–97. 2015.PubMed/NCBI

189 

Ryther RCC and Wong M: Mammalian target of rapamycin (mTOR) inhibition: Potential for antiseizure, antiepileptogenic, and epileptostatic therapy. Curr Neurol Neurosci Rep. 12:410–418. 2012.PubMed/NCBI

190 

Zeng LH, Xu L, Gutmann DH and Wong M: Rapamycin prevents epilepsy in a mouse model of tuberous sclerosis complex. Ann Neurol. 63:444–453. 2008.PubMed/NCBI

191 

Sunnen CN, Brewster AL, Lugo JN, Vanegas F, Turcios E, Mukhi S, Parghi D, D'Arcangelo G and Anderson AE: Inhibition of the mammalian target of rapamycin blocks epilepsy progression in NS-Pten conditional knockout mice. Epilepsia. 52:2065–2075. 2011.PubMed/NCBI

192 

Gericke B, Brandt C, Theilmann W, Welzel L, Schidlitzki A, Twele F, Kaczmarek E, Anjum M, Hillmann P and Löscher W: Selective inhibition of mTORC1/2 or PI3K/mTORC1/2 signaling does not prevent or modify epilepsy in the intrahippocampal kainate mouse model. Neuropharmacology. 162:1078172020.

193 

Buckmaster PS and Lew FH: Rapamycin suppresses mossy fiber sprouting but not seizure frequency in a mouse model of temporal lobe epilepsy. J Neurosci. 31:2337–2347. 2011.PubMed/NCBI

194 

Overwater IE, Rietman AB, van Eeghen AM and de Wit MCY: Everolimus for the treatment of refractory seizures associated with tuberous sclerosis complex (TSC): Current perspectives. Ther Clin Risk Manag. 15:951–955. 2019.PubMed/NCBI

195 

Franz DN, Lawson JA, Yapici Z, Ikeda H, Polster T, Nabbout R, Curatolo P, de Vries PJ, Dlugos DJ, Voi M, et al: Everolimus for treatment-refractory seizures in TSC: Extension of a randomized controlled trial. Neurol Clin Pract. 8:412–420. 2018.PubMed/NCBI

196 

Ravizza T, Scheper M, Di Sapia R, Gorter J, Aronica E and Vezzani A: mTOR and neuroinflammation in epilepsy: Implications for disease progression and treatment. Nat Rev Neurosci. 25:334–350. 2024.PubMed/NCBI

197 

Mazumder AG, Patial V and Singh D: Mycophenolate mofetil contributes to downregulation of the hippocampal interleukin type 2 and 1β mediated PI3K/AKT/mTOR pathway hyperactivation and attenuates neurobehavioral comorbidities in a rat model of temporal lobe epilepsy. Brain Behav Immun. 75:84–93. 2019.

198 

Drion CM, Borm LE, Kooijman L, Aronica E, Wadman WJ, Hartog AF, van Vliet EA and Gorter JA: Effects of rapamycin and curcumin treatment on the development of epilepsy after electrically induced status epilepticus in rats. Epilepsia. 57:688–697. 2016.PubMed/NCBI

199 

Brandt C, Hillmann P, Noack A, Römermann K, Öhler LA, Rageot D, Beaufils F, Melone A, Sele AM, Wymann MP, et al: The novel, catalytic mTORC1/2 inhibitor PQR620 and the PI3K/mTORC1/2 inhibitor PQR530 effectively cross the blood-brain barrier and increase seizure threshold in a mouse model of chronic epilepsy. Neuropharmacology. 140:107–120. 2018.PubMed/NCBI

200 

Castro OW, Upadhya D, Kodali M and Shetty AK: Resveratrol for easing status epilepticus induced brain injury, inflammation, epileptogenesis, and cognitive and memory Dysfunction-Are We There Yet? Front Neurol. 8:6032017.PubMed/NCBI

201 

Theilmann W, Gericke B, Schidlitzki A, Muneeb Anjum SM, Borsdorf S, Harries T, Roberds SL, Aguiar DJ, Brunner D, Leiser SC, et al: Novel brain permeant mTORC1/2 inhibitors are as efficacious as rapamycin or everolimus in mouse models of acquired partial epilepsy and tuberous sclerosis complex. Neuropharmacology. 180:1082972020. View Article : Google Scholar : PubMed/NCBI

202 

Wang Y and Chen Z: An update for epilepsy research and antiepileptic drug development: Toward precise circuit therapy. Pharmacol Ther. 201:77–93. 2019. View Article : Google Scholar : PubMed/NCBI

203 

Davis KL, Candrilli SD and Edin HM: Prevalence and cost of nonadherence with antiepileptic drugs in an adult managed care population. Epilepsia. 49:446–454. 2008. View Article : Google Scholar

204 

Klein P, Kaminski RM, Koepp M and Löscher W: New epilepsy therapies in development. Nat Rev Drug Discov. 23:682–708. 2024. View Article : Google Scholar : PubMed/NCBI

205 

Forcelli PA: Seizing control of neuronal activity: Chemogenetic applications in epilepsy. Epilepsy Curr. 22:303–308. 2022. View Article : Google Scholar : PubMed/NCBI

206 

Hadjiabadi D, Lovett-Barron M, Raikov IG, Sparks FT, Liao Z, Baraban SC, Leskovec J, Losonczy A, Deisseroth K and Soltesz I: Maximally selective single cell target for circuit control in epilepsy models. Neuron. 109:2556–2572.e6. 2021. View Article : Google Scholar :

207 

Löscher W: Drug Combinations for Antiepileptogenesis. Jasper's Basic Mechanisms Epilepsies. 1402–1418. 2024.

208 

Szklarczyk D, Santos A, Von Mering C, Jensen LJ, Bork P and Kuhn M: STITCH 5: Augmenting Protein-chemical interaction networks with tissue and affinity data. Nucleic Acids Res. 44:D380–D384. 2016. View Article : Google Scholar

209 

Welzel L, Bergin DH, Schidlitzki A, Twele F, Johne M, Klein P and Löscher W: Systematic evaluation of rationally chosen multitargeted drug combinations: A combination of low doses of levetiracetam, atorvastatin and ceftriaxone exerts antiepileptogenic effects in a mouse model of acquired epilepsy. Neurobiol Dis. 149:1052272021. View Article : Google Scholar

210 

Schidlitzki A, Bascuñana P, Srivastava PK, Welzel L, Twele F, Töllner K, Käufer C, Gericke B, Feleke R, Meier M, et al: Proof-of-concept that network pharmacology is effective to modify development of acquired temporal lobe epilepsy. Neurobiol Dis. 134:1046642020. View Article : Google Scholar

211 

Galanopoulou AS, Löscher W, Lubbers L, O'Brien TJ, Staley K, Vezzani A, D'Ambrosio R, White HS, Sontheimer H, Wolf JA, et al: Antiepileptogenesis and disease modification: Progress, challenges, and the path forward-Report of the Preclinical Working Group of the 2018 NINDS-sponsored antiepileptogenesis and disease modification workshop. Epilepsia Open. 6:276–296. 2021.PubMed/NCBI

212 

Porter RJ and Kupferberg HJ: The anticonvulsant screening program of the National institute of neurological disorders and stroke, NIH: History and contributions to clinical care in the twentieth century and beyond. Neurochem Res. 42:1889–1893. 2017.PubMed/NCBI

213 

Kehne JH, Klein BD, Raeissi S and Sharma S: The national institute of neurological disorders and stroke (NINDS) epilepsy therapy screening program (ETSP). Neurochem Res. 42:1894–1903. 2017.PubMed/NCBI

Related Articles

Journal Cover

May-2025
Volume 55 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dantio CD, Fasoranti DO, Teng C and Li X: Seizures in brain tumors: pathogenesis, risk factors and management (Review). Int J Mol Med 55: 82, 2025.
APA
Dantio, C.D., Fasoranti, D.O., Teng, C., & Li, X. (2025). Seizures in brain tumors: pathogenesis, risk factors and management (Review). International Journal of Molecular Medicine, 55, 82. https://doi.org/10.3892/ijmm.2025.5523
MLA
Dantio, C. D., Fasoranti, D. O., Teng, C., Li, X."Seizures in brain tumors: pathogenesis, risk factors and management (Review)". International Journal of Molecular Medicine 55.5 (2025): 82.
Chicago
Dantio, C. D., Fasoranti, D. O., Teng, C., Li, X."Seizures in brain tumors: pathogenesis, risk factors and management (Review)". International Journal of Molecular Medicine 55, no. 5 (2025): 82. https://doi.org/10.3892/ijmm.2025.5523