ABSTRACT
Seizures are a frequent complication in glioma. Incidence of brain tumor-related epilepsy (BTRE) in high-grade glioma (HGG) is an estimated > 25% and in low-grade glioma (LGG) is approximately 72%. Two first-line antiseizure medications (ASMs) for BTRE include levetiracetam (LEV) and valproic acid (VPA). Use of VPA has decreased because of a broader side effect profile, potential interaction with chemotherapeutic drugs, and availability of newer generation agents. In refractory BTRE, LEV and VPA may be prescribed together to enhance seizure control. VPA and LEV have gained attention for their purported antineoplastic effects and synergistic role with temozolomide. VPA is suggested to modulate anticancer activity in vitro through multiple mechanisms. In addition, retrospective studies indicate increased overall survival in patients with epileptogenic HGGs who are managed with LEV or VPA rather than other ASMs. However, these studies have numerous limitations. It is also reported that patients with glioma and a seizure history have a longer survival. This extended survival, if one exists, may be only observed in certain gliomas with corresponding patient characteristics. We provide a brief overview of the management of BTRE, VPA and LEV as anticonvulsants and antineoplastics, and the factors that may be associated with survival in epileptogenic glioma.
KEYWORDS: Brain tumor, epilepsy, levetiracetam, valproic acid, high-grade glioma, brain-tumor related epilepsy
1. Introduction
The clinical course of glioma is often complicated by seizures. Incidence of seizures is inversely related to the World Health Organization (WHO) grade of glioma [1]. While exact numbers are difficult to obtain, patients with high-grade glioma (HGG) have an incidence in the range of 25–60% [2], while low-grade glioma (LGG), mainly with an IDH1 mutation, have an incidence around 72% [3]. Numerous factors are involved in brain tumor-related epilepsy (BTRE), some of which include excitation-inhibition imbalance, structural and metabolic aberrations, genetic mutations, and immunological signals [4]. In addition to negatively [5] impacting quality of life, cortical hyperexcitability associated with seizures has been implicated [6,7] in glioma progression. Given that there may be a link between a hyperexcitable peritumoral environment in glioma and its disease burden, antiseizure medications (ASMs) have been suggested to confer a survival benefit in glioma [8].
Optimal management of brain-tumor related epilepsy (BTRE) is not fully codified. ASM prophylaxis is unwarranted for glioma patients who do not have a history of a seizure [9,10]. Patients with brain tumors are diagnosed with epilepsy in the event of their first seizure as there is a > 60% likelihood of additional seizures, meeting ILAE (International League Against Epilepsy) criteria for the diagnosis [11]. Although there has been limited data to provide high-level guidelines, BTRE is often managed with the same ASMs utilized for newly diagnosed nonlesional focal epilepsies. Current first-line monotherapies for adults with nonlesional focal epilepsy include but are not limited to: levetiracetam (LEV), lamotrigine, lacosamide, zonisamide, carbamazepine, and oxcarbazepine, with lamotrigine as a preferred option for first line when feasible [12–14]. ASMs favored as first-line for BTRE by both the Society for Neuro-Oncology (SNO) [15] and provider members of the European Association of Neuro-Oncology (EANO) [16] are lacosamide, lamotrigine, and LEV. Lamotrigine has the limitation of a slow titration schedule, taking up to 8–12 weeks for therapeutic dosing, limiting its use as a first-line agent in BTRE. Enzyme-inducing ASMs (EIASMs) are not recommended in the management of BTRE due to their modulation of the cytochrome P450 system and their potential impact on drug-drug interactions. Currently, ASM selection is irrespective of histology and localization of tumors [17]. Dosage titrations to maximum tolerated dose levels followed by alternate monotherapies or multidrug regimens are sought to achieve seizure control if patients are unresponsive to initial ASM treatment. Besides avoiding EIASMs, ASM preference depends on patient-specific factors. These include concurrent medications, drug-metabolizing enzyme polymorphisms, comorbidities, effective clearance (i.e., hepatic and kidney function), consideration of potential pregnancy, ease (or absence) of drug level monitoring, insurance coverage, and copays. Surgical resection of gliomas often has a benefit on seizures [18] as does minimally invasive stereotactic ablation [19] of epileptogenic tumors. In a subset of patients, management of BTRE remains challenging because it can be medically intractable [20]. In addition, there are unfavorable drug-drug interactions, and there is still incomplete insight into the pathophysiology of BTRE. Ultimately, the management of BTRE is driven by seizure control, quality of life, and survival [21].
LEV and VPA are two first-line ASMs utilized for BTRE [15–17,22]. At times, LEV-VPA dual therapy is used to manage refractory glioma-related epilepsy (GRE) [23]. There is support from a large multicenter retrospective study that these two agents in combination have the greatest overall efficacy in GRE, and their dual therapy may not be associated with more adverse effects compared to other ASM co-therapies [23]. However, this has not been validated in prospective trials. In addition to managing seizures, there are suggestions that the off-target activities of LEV and VPA may increase overall survival (OS) in glioma [8,24,25]. The majority of this data is based on retrospective studies [24]. The initially promising preliminary observations noted in these investigations have not been conclusively recapitulated in larger and more powerful investigations.
2. Mechanism of action
LEV is a pyrrolidine derivative utilized in the management of focal and generalized seizures [14]. Its main mechanism of action to decrease excitatory drive is blockade of synaptic vesicle protein 2A, which results in a reduction in the release of neurotransmitter vesicles (Figure 1) [26]. VPA is a branched short-chain fatty acid also used in the treatment of focal or generalized seizures in children and adults [14]. There are a few mechanisms by which VPA decreases neuronal excitation within the cortex (Figure 1). VPA increases the concentration of γ-aminobutyric acid (GABA) neurotransmission via inhibition of GABA transaminase [27]. Furthermore, VPA reduces epileptiform discharges through blockade of Na+ and Ca+ channels [28]. Given that LEV and VPA blunt neuronal activation through different mechanisms, they have demonstrated to be more effective together rather than independent agents in the management of intractable GRE [23].
Figure 1.

Mechanism of action levetiracetam blocks the release of neurotransmitter vesicles by inhibition of SV2A. Valproic acid increases the concentration of GABA via inhibition of GABAt. Valproic acid blocks Na+ and Ca2+ channels.
Abbreviations: AMPAr, AMPA receptor; GABA, -aminobutyrate; GABAt, GABA transaminase; NMDAr, N-methyl-Daspartate receptor; SV2A, synaptic vesicle protein 2A.
3. Monotherapy – levetiracetam
LEV is a first-line agent and likely the most common ASM used in the management of BTRE. It carries the following pharmacokinetic advantages: absence of cytochrome P450 modulation, low potential of drug-drug interactions, good bioavailability in IV formulations, rapid titration to an effective dose, and relative safety at pregnancy [29,30]. Additionally, LEV is suitable for a wide patient population in the United States because it is available in liquid formulations and is offered in a generic prescription. LEV may be associated with some of the highest rates of seizure freedom compared to other ASMs in GRE, with a 74% six-month seizure-free weighted average [31]. Furthermore, LEV has been associated with the highest likelihood of achieving a ≥ 50% reduction in seizure frequency and demonstrated the lowest treatment failure among other ASMs [31]. In a comparative study, LEV demonstrated similar efficacy as phenytoin in managing GRE, but LEV was preferred because of less seizure-unrelated dose adjustments and a more favorable side-effect profile [32]. After 2 years of treatment, the rates of seizure freedom (i.e., no seizures for 12 months) in WHO grade 2 GRE appears best achieved with LEV compared to other ASMs [33]. At this time, the ASM provided with the greatest support in the management of BTRE is LEV.
The most concerning side effect recognized with LEV is neuropsychiatric disturbance (e.g., emotional lability, anxiety, depression) [34,35]. These mood disturbances associated with LEV are most common when brain tumors are located in the frontal lobe [36]. Therefore, as a treatment option for BTRE, LEV can be impacted by the dose-limiting toxicity of irritability observed in a subset of patients. This is of importance, as many glioma patients are treated concurrently with steroids, which may further heighten the neuropsychiatric side effects of LEV. It is essential for clinicians to monitor for mood changes, depression, and irritability. The side effect profile of LEV is summarized in Table 1.
Table 1.
Side Effect Profile.
| Side Effect Profile | |
|---|---|
| Levetiracetam [29,37,40] | Somnolence, fatigue, infection, dizziness, anxiety, depression, aggression, agitation |
| Valproic Acid [41,45] | Weight gain, hair loss, polycystic ovary, pancreatitis, gastrointestinal disturbances, hepatoxicity, encephalopathy, coagulopathy, teratogenicity, tremor |
4. Monotherapy - valproic acid
Focal seizures with or without secondary generalization are the most common manifestation of epileptogenic glioma [38,39]. VPA is a reasonable option for BTRE, given that it is has level B evidence for nonlesional focal epilepsies [14]. In addition to epilepsy management, VPA is also approved for the management of bipolar disorder and migraine prophylaxis. In an EANO survey of mostly European physicians, 21% chose VPA as a first-line alternative to LEV in managing BTRE [16]. Alternative options for the management of BTRE were not queried in another survey of American surgeons, but 85% selected LEV and 0% selected VPA as a first-line agent for postoperative prophylaxis [40]. Compared to LEV, VPA has been shown to result in more treatment failures in WHO grade 2–4 GRE 12 months after ASM initiation (50% vs 33%) [41]. Nonetheless, VPA has demonstrated efficacy in GRE in other studies. In a cohort of patients with glioblastoma (GBM), VPA and LEV had comparable seizure freedom at a follow-up ≥6 months (41% and 43%) [42]. At 6 months after surgery, VPA achieved postoperative seizure control in 61% of WHO grade 2 GRE [43]. Even though VPA attains efficacy for the management of GRE, it has fallen out of favor over LEV and newer generation ASMs, which might be directly related to its side effect profile.
VPA has a broad side effect profile, with the ability to cause injury across multiple organs. VPA may lead to neurological (e.g., tremor), hematological (e.g., thrombocytopenia), and gastrointestinal (e.g., nausea, constipation) symptoms [44]. Albeit uncommon, life-threatening toxicities are associated with VPA (e.g., cutaneous hypersensitivity reactions, hepatitis, and pancreatitis) [45]. Due to the incidence of transaminitis (15%) [46], hematologic abnormalities (33%) [47], and hyperammonemia (28%) [48], routine lab monitoring should be considered. A baseline complete blood count with differential and liver function tests should be performed with subsequent routine monitoring, especially within the first 6 months of treatment. The majority of adverse effects of VPA are often controlled with appropriate dosage adjustments. Importantly, VPA is a cytochrome P450 enzyme inhibitor; therefore, VPA may lead to drug-drug interactions and exclude patients from clinical drug trials [49]. Valproic acid should be avoided in women of childbearing age as it carries the highest risk of teratogenic effects including risk of neural tube defects [50], lower intelligence quotient [13], neurodevelopmental disorders (i.e., autism) [51] in offspring. The side effects of VPA are summarized in Table 1.
5. Dual therapy - levetiracetam & valproic acid
GRE may be refractory to ASMs [20,52,53]. In these cases, a trend toward an add-on ASM is the choice of management rather than sequential monotherapy trials. LEV-VPA combinatorial therapy has been shown to enhance seizure control for GRE compared to each of their stand-alone managements [23]. This is most likely because VPA and LEV mitigate cortical hyperexcitability through different mechanisms. A SNO consensus recommends LEV-VPA as a potential treatment option for BTRE if a first-line agent is ineffective [15]. Among dual ASM combinations, the value of LEV-VPA for intractable BTRE is documented in a few reports [23,42,54]. In a cohort of various brain neoplasms, LEV-VPA dual therapy, unsurprisingly, provided better seizure control compared to VPA or LEV monotherapies after ≥6 months of treatment [54]. Among patients with WHO grade 2–4 GRE, a large multicenter analysis further distinguished LEV-VPA dual therapy to provide superior seizure control than other alternative dual therapy combinations that included either VPA or LEV in their regimen (e.g., clobazam, phenytoin, lacosamide, etc.) [23]. Furthermore, in a study of GBM-related epilepsy, a subset of patients continued to have seizures with VPA or LEV. Among them, 54% achieved seizure freedom after LEV-VPA co-therapy [42]. Given their different mechanisms of action and combined tolerability, LEV-VPA dual therapy is a suitable, but certainly not the only, next-step treatment option for intractable BTRE.
6. Thrombocytopenia - temozolomide & valproic acid
Therapy with either temozolomide (TMZ) or VPA can lead to low blood cell counts, bringing into question if there is an increased risk of cytopenia for patients treated simultaneously with both of these agents [55,56]. In particular, platelet levels are vulnerable to decline with TMZ or VPA treatment [47,57]. Although the exact mechanism is unknown, VPA is thought to peripherally destroy platelets [47]. Myelosuppression is often seen with TMZ because of the low expression of MGMT in the bone marrow [58]. In matched cohorts of GRE (±TMZ) managed with either VPA (n = 429) or LEV (n = 429), ASM failure rates from adverse events were comparable among the two agents at 16% for LEV and 17% for VPA. However, low platelet counts led to 14% of all discontinuations of VPA. Ten patients (2.3%) developed grade 1/2 and 6 patients (1.3%) developed grade 3/4 thrombocytopenia [40]. In a clinical trial subanalysis of newly diagnosed GBM patients receiving chemoradiotherapy, VPA led to grade 3 or 4 thrombocytopenia. Patients had delay in their TMZ treatment, but ultimately, they did not have more dose reductions [59]. In a retrospective study of GBM patients treated with TMZ-VPA, TMZ was the only variable that reached the platelet threshold (<100,000/mm3) for TMZ dosage modification [60]. During and 4–8 weeks after chemoradiotherapy, relative platelet stability has been observed in VPA compared to LEV or no prescribed ASM. Levels of neutrophils, lymphocytes and thrombocytes decreased unanimously across these three treatment groups [61]. It is worthy to mention, in addition to TMZ and VPA, radiation that is a part of the standard treatment of HGG may contribute to hematological toxicity [62]. In the aggregate, in contrast to other ASMs, VPA has a slightly increased risk for thrombocytopenia in patients with HGG who are prescribed concurrent TMZ treatment. Therefore, routine hematological surveillance should be performed to monitor patients if VPA is chosen as agent to manage GRE.
7. Seizures & overall survival
Although perhaps counterintuitive, a preoperative seizure has been associated with prolonged OS in glioma, and this association has been most described in WHO grade 3–4 glioma [63–66]. About one-fourth of patients are diagnosed with GBM after they present with a seizure as their initial symptom [38,67,68]. In a retrospective nationwide study, mortality risk was lower in patients with a seizure <1 year prior to WHO grade 2–4 glioma diagnosis [69]. A notable limitation of many studies in this space is the incorporation of both isocitrate dehydrogenase (IDH)-wild-type (older age, unfavorable prognosis, lower seizure incidence) and IDH-mutant (younger [70], favorable [71], higher [3]) glioma patient populations into single cohorts. Moving forward, studies exclusively analyzing patients based on the 2021 WHO Classification of CNS Tumors [72], which codified adult infiltrating glioma classification on the IDH mutational status, will hopefully overcome this substantial limitation.
Patients with HGG are susceptible to a delay in diagnosis when presenting with nonspecific signs (e.g., headache, weakness, fatigue) [73]. In the event of an abrupt, unprovoked seizure, HGG patients may be more inclined to seek medical attention (e.g., present to an emergency room) and receive brain imaging. Thus, glioma patients with a seizure may present in an earlier stage of disease [74] and may receive earlier treatment [75]. On the other hand, an initial seizure may simply cause a diagnostic lead-time bias and skew longer OS. Contrast-enhanced MRI of newly diagnosed GBM have also shown a distinctive microstructure for epileptogenic tumors which might indicate a less aggressive growth pattern [76]. Volumetric analyses measure epileptogenic HGGs smaller than their non-epileptogenic analog [68,76–79]. In turn, a smaller tumor increases the probability to achieve a greater extent of resection, a robust prognostic marker [80]. Moreover, epileptogenic glioma may be surgically advantageous, given their more accessible location at the superficial cortex [78]. Patients presenting with seizures are also younger at the time of primary glioma resection [1,37,78,79,81,82]. An increased duration of functional independence (a correlate to extended OS) is furthermore observed with a preoperative seizure in patients with GBM after surgery [83].
Other reports have not found any association between longer OS and seizures in GBM [79,84,85]. The prognosis of GBM and seizures seems to be impacted differently across age groups, with older patients demonstrating decreased survival with epilepsy [86]. The general comorbidities associated with seizures (e.g., stroke) can also lead to worsened survival [87]. Lastly, preclinical data have expounded on the association of neuronal excitability and malignancy in glioma [6,7]. The direct synaptic connection between neurons and glioma has been implicated to drive glioma progression and invasiveness [6,7]. There are a multitude of variables to take into consideration while evaluating the effect of seizures on glioma survival. Currently, more data is required to determine whether there is a connection between epileptogenic glioma and OS, but this outcome appears to be different between specific subgroups of patients.
8. Prolonged survival
TMZ, a DNA methylating agent, is a part of the standard management of malignant glioma [88]. LEV is thought to hold value in the management of HGG, in part due to its purported bioaction to augment the apoptotic effects of TMZ via O6-methylguanine-DNA methyltransferase (MGMT) transcriptional inhibition in vitro [89–91]. Single-institutional retrospective studies have shown prolonged median OS with TMZ-LEV dual therapy versus TMZ monotherapy [92–95]. The impact of these studies is limited by their intrinsic design. A single institution retrospectively case-matched GBM patients (n = 460) who either received continuous, intermittent or no LEV during standard chemotherapy. The investigators found that continuous LEV treatment was associated with longer OS compared to the two other cohorts [95]. Perioperative administration of LEV has been correlated with increased progression-free survival (PFS) and OS [96]. In an open-label single-arm phase 2 study, LEV was associated with prolonged GBM survival in younger patients and those with unmethylated MGMT promoter status [97]. These studies indicating increased survival with LEV have several methodological limitations. As an example, in one study, patients who had extended survival with LEV were diagnosed with HGG after a seizure and a higher percentage of patients underwent complete resection [92]. The survival benefit of LEV treatment has not been observed in other studies [98–100]. A double-blind, randomized control trial is ongoing to determine the survival outcome of TMZ-LEV treatment in Chinese GBM patients [101]. Before impacting clinical practice, more data is needed to validate LEV favorably impacts OS.
The finding that VPA behaves as a histone deacetylase (HDAC) inhibitor has also implicated its potential use as an antitumor agent [102]. In addition to this mechanism, there are various other modalities by which VPA might facilitate an antitumor effect. VPA is reported to mitigate tumoral proliferation [103], invasion [104], and angiogenesis [105], in addition to attaining a synergistic [106–108] effect with the antineoplastic effects of TMZ in several glioma cell cultures (Figure 2). It also demonstrates a radio-sensitizing [109,110] and radio-protecting [109] effect upon GBM cells and healthy cells, respectively. It is possible that the benefit of HDAC inhibition may be most pronounced in the IDH-mutant glioma population, where this approach has not been as extensively studied [111]. Retrospective studies initially suggested that VPA co-medication during chemoradiation extends OS [42,59,112,113]. This observation was supported by systematic reviews and meta-analyses [24,114,115]. However, an increase in OS has not been recapitulated in more recent studies [24]. It is possible enhanced survival with VPA can be confounded by age. In a nationwide analysis, TMZ-VPA treatment prolonged OS to the greatest extent in younger patients [116]. Lastly, after pooling data from four randomized control trials, there was no improvement of OS and PFS in newly diagnosed GBM patients when they were treated with VPA (or LEV), indicating that ASMs should only be used to manage seizures and not malignancy [117].
Figure 2.

Levetiracetam and valproic acid attain a synergistic effect with temozolomide in cell culture studies. Valproic acid has shown anti-glioma activity in vitro through mitigation of cellular proliferation, tumor invasion and angiogenesis. Valproic acid induces radiosensitization of tumor cells and inhibits HDAC.
Abbreviations: HDAC, histone deacetylase.
9. Future perspective
Randomized clinical trials are one means to evaluate both the anticonvulsant efficacies and antineoplastic properties of LEV and VPA. However, as resources are limited, and the expectation is modest for a marked clinical impact with the addition of an ASM to standard of care treatments, the likelihood of such trials moving forward and completing enrollment is somewhat low [118]. These trials, if conducted, could provide data regarding seizure control, toxicities, and survival outcomes of LEV and VPA. Analyses of such trials should separate tumors based on molecular markers. IDH is one example, as an increased risk of seizures and increased OS is observed with this mutational status [3]. D-2-Hydroxyglutarate, an oncometabolite produced in IDH-mutant glioma, has shown to contribute to this seizure activity [119], and IDH inhibitors appear to decrease seizure burden [120]. Stratification of glioma based on tumor grade is also insightful, given that the tumor grade is inversely related to seizure incidence [1]. Furthermore, there is uncertainty to whether LEV or VPA penetrate malignant glia to produce a significant tumoricidal environment in vivo. The appropriate dosages, if a difference exists, for seizure control compared to anti-glioma activity needs to be determined for these agents. Phase 0 clinical trials can help evaluate these queries by measuring intratumoral concentrations of LEV and VPA that is administered prior to surgical resection. While such trials have been proposed, to our knowledge, none are actively accruing patients at this time.
10. Conclusion
LEV and VPA are two commonly used agents for seizure control in glioma. They are both categorized as first-line agents, but VPA is more often utilized as an add-on or alternative agent when a monotherapy is ineffective. LEV and VPA have demonstrated antineoplastic effects in preclinical studies. In addition, some retrospective studies have also suggested increased survival with these agents. At this time, the proposal that LEV and VPA increases glioma survival and incorporation of these agents into clinical practice for this reason has not been validated in randomized trials and the current evidence is not strong enough to recommend clinical use beyond antiseizure activity. The majority of studies supporting prolonged survival are utilizing data extrapolated from retrospective studies where compliance and dosage of LEV and VPA were not quantified. In addition, the extended survival observed with these agents has been most pronounced in the younger population [97,116]. VPA is suggested to slow malignant progression in preclinical studies via inhibition of HDAC. Previous clinical studies investigating HDAC inhibition (e.g., vorinostat, romidepsin) as a therapeutic option for GBM have been ineffective [121,122]. In comparison to these trialed agents, VPA is a less potent HDAC inhibitor, and it appears therefore less probable VPA is able to slow glioma progression through this mechanism. Nonetheless, numerous modalities have been described by which VPA exerts its purported antineoplastic effects [123]. The association of seizures and glioma survival is a complex topic with heterogeneous findings. Survival is most likely influenced by a multitude of factors, including but not limited to tumor grade, molecular subtype, transcriptional modification (e.g., MGMT methylation) and baseline cohort characteristics (e.g., age, functional status). The idea of repurposing ASMs as an adjuvant agent for glioma progression will require further investigation through clinical trials prior to impacting the clinical practice.
Funding Statement
R.V.L has received funding from P50CA221747 and BrainUp.
Disclosure statement
R.V.L has received research support (drug only) from BMS, honoraria for serving on Advisory Boards for AstraZeneca, Cardinal Health, Curio, Merck, Novartis, Novocure, Servier, and Telix, honoraria for consulting for Novartis and Servier, honoraria for serving on the Speakers’ Bureau for Merck, Novocure, and Servier, and honoraria for editing for EBSCO, Elsevier, Medlink Neurology, and Oxford University Press. T.W. is on the Advisory Board of Alexion, Springworks, Servier and the Data Monitoring Board of Novocure, IQVIA. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Article highlights
Two first-line agents utilized in the management of glioma-related epilepsy include levetiracetam and valproic acid.
Beyond seizure control, an extended survival via anticancer mechanisms have been attributed to both levetiracetam and valproic acid.
Levetiracetam and valproic acid in combination may increase seizure freedom rates in patients with refractory glioma-related epilepsy.
Seizures are believed to influence survival in glioma; although, these findings are heterogeneous across tumor and patient characteristics.
Prospective studies are needed to validate whether antiseizure medications have a role in the oncologic management of glioma.
Author contributions
BFK, RVL, and JWT conceptualized the content and structure of the review. BFK wrote the initial draft of the manuscript. BFK, TW, KD, CH, KG, HT, MCT, RS, RVL, and JWT extensively revised the manuscript for intellectual content. All authors approved the final draft.
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
- 1.Li L, Fang S, Li G, et al. Glioma-related epilepsy in patients with diffuse high-grade glioma after the 2016 WHO update: seizure characteristics, risk factors, and clinical outcomes. J Neurosurg. 2021;136(1):67–75. doi: 10.3171/2020.12.JNS203351 [DOI] [PubMed] [Google Scholar]
- 2.Englot DJ, Berger MS, Chang EF, et al. Characteristics and treatment of seizures in patients with high-grade glioma: a review. Neurosurg Clinics NA. 2012;23:227–235. doi: 10.1016/j.nec.2012.01.009 [DOI] [PubMed] [Google Scholar]
- 3.Li Y, Shan X, Wu Z, et al. IDH1 mutation is associated with a higher preoperative seizure incidence in low-grade glioma: a systematic review and meta-analysis. Seizure. 2018;55:76–82. doi: 10.1016/J.SEIZURE.2018.01.011 [DOI] [PubMed] [Google Scholar]
- 4.Hills KE, Kostarelos K, Wykes RC.. Converging mechanisms of Epileptogenesis and their insight in glioblastoma. Front Mol Neurosci. 2022. 15. doi: 10.3389/FNMOL.2022.903115 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Review of the pathophysiology of brain-tumor related epilepsy in glioblastoma.
- 5.Shin JY, Kizilbash SH, Robinson SI, et al. Seizures in patients with primary brain tumors: what is their psychosocial impact? J Neurooncol. 2016;128(2):285–291. doi: 10.1007/S11060-016-2108-Y/TABLES/2 [DOI] [PubMed] [Google Scholar]
- 6.Venkatesh HS, Morishita W, Geraghty AC, et al. Electrical and synaptic integration of glioma into neural circuits. Nature. 2019;573(7775):539–545. doi: 10.1038/S41586-019-1563-Y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Venkataramani V, Tanev DI, Strahle C, et al. Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature. 2019;573(7775):532–538. doi: 10.1038/s41586-019-1564-x [DOI] [PubMed] [Google Scholar]
- 8.Rilinger RG, Guo L, Sharma A, et al. Tumor-related epilepsy in high-grade glioma: a large series survival analysis. J Neurooncol. Published online 2024. doi: 10.1007/S11060-024-04787-Z [DOI] [PMC free article] [PubMed] [Google Scholar]; • Retrospective review that found seizures lead to better survival in glioblastoma irrespective of IDH mutational status.
- 9.Walbert T, Harrison RA, Schiff D, et al. SNO and EANO practice guideline update: anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Neuro Oncol. 2021;23(11):1835–1844. doi: 10.1093/NEUONC/NOAB152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chen CC, Rennert RC, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines on the role of prophylactic Anticonvulsants in the treatment of adults with metastatic brain tumors. Clin Neurosurg. 2019;84(3):E195–E197. doi: 10.1093/NEUROS/NYY545 [DOI] [PubMed] [Google Scholar]
- 11.Fisher RS, Acevedo C, Arzimanoglou A, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55(4):475–482. doi: 10.1111/EPI.12550 [DOI] [PubMed] [Google Scholar]
- 12.Marson A, Burnside G, Appleton R, et al. The SANAD II study of the effectiveness and cost-effectiveness of levetiracetam, zonisamide, or lamotrigine for newly diagnosed focal epilepsy: an open-label, non-inferiority, multicentre, phase 4, randomised controlled trial. Lancet. 2021;397(10282):1363–1374. doi: 10.1016/S0140-6736(21)00247-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kanner AM, Bicchi MM. Antiseizure medications for adults with epilepsy: a review. JAMA. 2022;327(13):1269–1281. doi: 10.1001/JAMA.2022.3880 [DOI] [PubMed] [Google Scholar]
- 14.Glauser T, Ben-Menachem E, Bourgeois B, et al. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 2013;54(3):551–563. doi: 10.1111/EPI.12074 [DOI] [PubMed] [Google Scholar]
- 15.Avila EK, Tobochnik S, Inati SK, et al. Brain tumor-related epilepsy management: a society for neuro-oncology (SNO) consensus review on current management. Neuro Oncol. 2024;26(1). doi: 10.1093/NEUONC/NOAD154 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Recommendations for management of brain-tumor related epilepsy detailed by the Society for Neuro-oncology.
- 16.Van Der Meer PB, Dirven L, Van Den Bent MJ, et al. Prescription preferences of antiepileptic drugs in brain tumor patients: an international survey among EANO members. Neurooncol Pract. 2021;9(2):105–113. doi: 10.1093/NOP/NPAB059 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Survey of provider members of the European Association of Neuro-Oncology regarding their antiseizure medications of choice.
- 17.Maschio M, Aguglia U, Avanzini G, et al. Management of epilepsy in brain tumors. Neurological Sci. 2019;40(10):2217–2234. doi: 10.1007/S10072-019-04025-9 [DOI] [PubMed] [Google Scholar]
- 18.Englot DJ, Berger MS, Barbaro NM, et al. Predictors of seizure freedom after resection of supratentorial low-grade gliomas: a review. J Neurosurg. 2011;115(2):240–244. doi: 10.3171/2011.3.JNS1153 [DOI] [PubMed] [Google Scholar]
- 19.Nielsen SH, Rasmussen R. Mr-guided laser interstitial thermal therapy in the treatment of brain tumors and epilepsy. Acta Neurochir (Wien). 2024;166(1):1–11. doi: 10.1007/S00701-024-06238-0/METRICS [DOI] [PubMed] [Google Scholar]
- 20.Van Der Meer PB, Dirven L, Fiocco M, et al. Effectiveness of antiseizure medication triple therapy in patients with glioma with refractory epilepsy: an observational cohort study. Neurology. 2023;100(14):E1488–E1496. doi: 10.1212/WNL.0000000000206852/ASSET/2DD055A0-23C4-4656-B276-61663F2B99FB/ASSETS/IMAGES/LARGE/8TTU1.JPG [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Nowell M, Miserocchi A, McEvoy AW. Tumors in epilepsy. Semin Neurol. 2015;35(3):209–217. doi: 10.1055/S-0035-1552628 [DOI] [PubMed] [Google Scholar]
- 22.Sokolov E, Dietrich J, Cole AJ. The complexities underlying epilepsy in people with glioblastoma. Lancet Neurol. 2023;22(6):505–516. doi: 10.1016/S1474-4422(23)00031-5 [DOI] [PubMed] [Google Scholar]; •• Review of the pathophysiology, management, and antineoplastic activities of antiseizure medications in glioblastoma-related epilepsy.
- 23.Van Der Meer PB, Dirven L, Fiocco M, et al. Effectiveness of antiseizure medication duotherapies in patients with glioma: a multicenter observational cohort study. Neurology. 2022;99(10):E999–E1008. doi: 10.1212/WNL.0000000000200807/ASSET/7DFBA8EA-6B86-4A1A-AA53-9D3755988F4F/ASSETS/GRAPHIC/20TTU1.JPEG [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Retrospective review of the efficacies and side effect profiles of valproic acid and levetiracetam in matched cohorts of WHO grade 2–4 glioma-related epilepsy.
- 24.Lu VM, Texakalidis P, McDonald KL, et al. The survival effect of valproic acid in glioblastoma and its current trend: a systematic review and meta-analysis. Clin Neurol Neurosurg. 2018;174:149–155. doi: 10.1016/J.CLINEURO.2018.09.019 [DOI] [PubMed] [Google Scholar]
- 25.Fan X, Li Y, Shan X, et al. Seizures at presentation are correlated with better survival outcomes in adult diffuse glioma: a systematic review and meta-analysis. Seizure. 2018;59:16–23. doi: 10.1016/J.SEIZURE.2018.04.018 [DOI] [PubMed] [Google Scholar]
- 26.Lynch BA, Lambeng N, Nocka K, et al. The synaptic vesicle is the protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc Natl Acad Sci USA. 2004;101(26):9861–9866. doi: 10.1073/PNAS.0308208101/ASSET/329D471F-1001-41CF-89A6-C93AD65511F9/ASSETS/GRAPHIC/ZPQ0260452550006.JPEG [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Johannessen CU, Johannessen SI. Valproate: past, present, and future. CNS Drug Rev. 2003;9(2):199–216. doi: 10.1111/J.1527-3458.2003.TB00249.X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Romoli M, Mazzocchetti P, D’Alonzo R, et al. Valproic acid and Epilepsy: from molecular mechanisms to clinical evidences. Curr Neuropharmacol. 2019;17(10):926. doi: 10.2174/1570159X17666181227165722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Abou-Khalil B. Levetiracetam in the treatment of epilepsy. Neuropsychiatr Dis Treat. 2008;4(3):507–523. doi: 10.2147/NDT.S2937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mawhinney E, Craig J, Morrow J, et al. Levetiracetam in pregnancy: results from the UK and Ireland epilepsy and pregnancy registers. Neurology. 2013;80(4):400–405. doi: 10.1212/WNL.0B013E31827F0874/SUPPL_FILE/MAWHINNEY_400.PDF [DOI] [PubMed] [Google Scholar]
- 31.De Bruin ME, Van Der Meer PB, Dirven L, et al. Efficacy of antiepileptic drugs in glioma patients with epilepsy: a systematic review. Neurooncol Pract. 2021;8(5):501–517. doi: 10.1093/NOP/NPAB030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Merrell RT, Anderson SK, Meyer FB, et al. Seizures in patients with glioma treated with phenytoin and levetiracetam. J Neurosurg. 2010;113(6):1176–1181. doi: 10.3171/2010.5.JNS091367 [DOI] [PubMed] [Google Scholar]
- 33.Fairclough S, Goodden J, Chumas P, et al. Levetiracetam as a first-line antiseizure medication in WHO grade 2 glioma: time to seizure freedom and rates of treatment failure. Epilepsia. 2023;64(4):857–865. doi: 10.1111/EPI.17508 [DOI] [PubMed] [Google Scholar]
- 34.Seystahl K, Oppong FB, Le Rhun E, et al. Associations of levetiracetam use with the safety and tolerability profile of chemoradiotherapy for patients with newly diagnosed glioblastoma. Neurooncol Adv. 2022;4(1):1–13. doi: 10.1093/NOAJNL/VDAC112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Harden C. Safety profile of levetiracetam. Epilepsia. 2001;42(SUPPL. 4):36–39. doi: 10.1111/J.1528-1167.2001.00008.X [DOI] [PubMed] [Google Scholar]
- 36.Bedetti C, Romoli M, Maschio M, et al. Neuropsychiatric adverse events of antiepileptic drugs in brain tumour-related epilepsy: an Italian multicentre prospective observational study. Eur J Neurol. 2017;24:1283–1289. doi: 10.1111/ene.13375 [DOI] [PubMed] [Google Scholar]
- 37.French J, Edrich P, Cramer JA. A systematic review of the safety profile of levetiracetam: a new antiepileptic drug. Epilepsy Res. 2001;47(1–2):77–90. doi: 10.1016/S0920-1211(01)00296-0 [DOI] [PubMed] [Google Scholar]
- 38.Toledo M, Sarria-Estrada S, Quintana M, et al. Epileptic features and survival in glioblastomas presenting with seizures. Epilepsy Res. 2017;130:1–6. doi: 10.1016/J.EPLEPSYRES.2016.12.013 [DOI] [PubMed] [Google Scholar]
- 39.Kerkhof M, Vecht CJ. Seizure characteristics and prognostic factors of gliomas. Epilepsia. 2013;54 Suppl 9(SUPPL. 9):12–17. doi: 10.1111/EPI.12437 [DOI] [PubMed] [Google Scholar]
- 40.Dewan MC, Thompson RC, Kalkanis SN, et al. Prophylactic antiepileptic drug administration following brain tumor resection: results of a recent AANS/CNS section on tumors survey. J Neurosurg. 2016;126(6):1772–1778. doi: 10.3171/2016.4.JNS16245 [DOI] [PubMed] [Google Scholar]
- 41.van der Meer PB, Dirven L, Fiocco M, et al. First‐line antiepileptic drug treatment in glioma patients with epilepsy: levetiracetam vs valproic acid. Epilepsia. 2021;62(5):1119. doi: 10.1111/EPI.16880 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kerkhof M, Dielemans JCM, Van Breemen MS, et al. Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro Oncol. 2013;15(7):961–967. doi: 10.1093/NEUONC/NOT057 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.You G, Sha ZY, Yan W, et al. Seizure characteristics and outcomes in 508 Chinese adult patients undergoing primary resection of low-grade gliomas: a clinicopathological study. doi: 10.1093/neuonc/nor205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nanau RM, Neuman MG. Adverse drug reactions induced by valproic acid. Clin Biochem. 2013;46(15):1323–1338. doi: 10.1016/J.CLINBIOCHEM.2013.06.012 [DOI] [PubMed] [Google Scholar]
- 45.Gerstner T, Bell N, König S. Oral valproic acid for epilepsy–long-term experience in therapy and side effects. Expert Opin Pharmacother. 2008;9(2):285–292. doi: 10.1517/14656566.9.2.285 [DOI] [PubMed] [Google Scholar]
- 46.Cotariu D, Zaidman JL. Valproic acid and the liver. Clin Chem. 1988;34(5):890–897. doi: 10.1093/CLINCHEM/34.5.890 [DOI] [PubMed] [Google Scholar]
- 47.Acharya S, Bussel JB. Hematologic toxicity of sodium valproate. J Pediatr Hematol Oncol. 2000;22(1):62–65. doi: 10.1097/00043426-200001000-00012 [DOI] [PubMed] [Google Scholar]
- 48.Tseng YL, Huang CR, Lin CH, et al. Risk factors of hyperammonemia in patients with epilepsy under valproic acid therapy. Medicine (Baltimore). 2014;93(11):e66. doi: 10.1097/MD.0000000000000066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhu MM, Li HL, Shi LH, et al. The pharmacogenomics of valproic acid. J Hum Genet. 2017;62(12):1009–1014. doi: 10.1038/JHG.2017.91 [DOI] [PubMed] [Google Scholar]
- 50.Nau H. Valproic acid-induced neural tube defects. Ciba Found Symp. 1994;181:144–160. doi: 10.1002/9780470514559.CH9 [DOI] [PubMed] [Google Scholar]
- 51.Christensen J, Grnøborg TK, Srøensen MJ, et al. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309(16):1696–1703. doi: 10.1001/JAMA.2013.2270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Jo J, Nevel K, Sutyla R, et al. Predictors of early, recurrent, and intractable seizures in low-grade glioma. Neurooncol Pract. 2020;8(1):40–47. doi: 10.1093/NOP/NPAA054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yao PS, Zheng SF, Wang F, et al. Surgery guided with intraoperative electrocorticography in patients with low-grade glioma and refractory seizures. J Neurosurg. 2017;128(3):840–845. doi: 10.3171/2016.11.JNS161296 [DOI] [PubMed] [Google Scholar]
- 54.Van Breemen MSM, Rijsman RM, Taphoorn MJB, et al. Efficacy of anti-epileptic drugs in patients with gliomas and seizures. J Neurol. 2009;256(9):1519–1526. doi: 10.1007/S00415-009-5156-9 [DOI] [PubMed] [Google Scholar]
- 55.Scaringi C, De Sanctis V, Minniti G, et al. Temozolomide-related hematologic toxicity. Onkologie. 2013;36(7–8):444–449. doi: 10.1159/000353752 [DOI] [PubMed] [Google Scholar]
- 56.Buoli M, Serati M, Botturi A, et al. The risk of thrombocytopenia during valproic acid therapy: a critical summary of available clinical data. Drugs R D. 2018;18(1):1–5. doi: 10.1007/S40268-017-0224-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Gerber DE, Grossman SA, Zeltzman M, et al. The impact of thrombocytopenia from temozolomide and radiation in newly diagnosed adults with high-grade gliomas. Neuro Oncol. 2007;9(1):47. doi: 10.1215/15228517-2006-024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hansen WK, Kelley MR. Review of mammalian DNA repair and translational implications 1. Published online 2000 [cited 2024 Aug 16]. Available from: http://www.jpet.org [PubMed]
- 59.Weller M, Gorlia T, Cairncross JG, et al. Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma. Neurology. 2011;77(12):1156–1164. doi: 10.1212/WNL.0B013E31822F02E1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Simó M, Velasco R, Graus F, et al. Impact of antiepileptic drugs on thrombocytopenia in glioblastoma patients treated with standard chemoradiotherapy. J Neurooncol. 2012;108(3):451–458. doi: 10.1007/S11060-012-0836-1 [DOI] [PubMed] [Google Scholar]
- 61.Tinchon A, Oberndorfer S, Marosi C, et al. Haematological toxicity of valproic acid compared to Levetiracetam in patients with glioblastoma multiforme undergoing concomitant radio-chemotherapy: a retrospective cohort study. J Neurol. 2015;262(1):179–186. doi: 10.1007/S00415-014-7552-Z [DOI] [PubMed] [Google Scholar]
- 62.Jefferies S, Rajah B, Ashley S, et al. Haematological toxicity of cranio-spinal irradiation. Radiother Oncol. 1998;48(1):23–27. doi: 10.1016/S0167-8140(98)00024-3 [DOI] [PubMed] [Google Scholar]
- 63.Toledo M, Sarria-Estrada S, Quintana M, et al. Prognostic implications of epilepsy in glioblastomas. Clin Neurol Neurosurg. 2015;139:166–171. doi: 10.1016/J.CLINEURO.2015.10.002 [DOI] [PubMed] [Google Scholar]
- 64.Yang P, You G, Zhang W, et al. Correlation of preoperative seizures with clinicopathological factors and prognosis in anaplastic gliomas: a report of 198 patients from China. Seizure. 2014;23(10):844–851. doi: 10.1016/J.SEIZURE.2014.07.003 [DOI] [PubMed] [Google Scholar]
- 65.Berendsen S, Varkila M, Kroonen J, et al. Prognostic relevance of epilepsy at presentation in glioblastoma patients. Neuro Oncol. 2016;18(5):700. doi: 10.1093/NEUONC/NOV238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ge H, Di G, Yan Z, et al. Does epilepsy always indicate worse outcomes? A longitudinal follow-up analysis of 485 glioma patients. World J Surg Oncol. 2022;20(1). doi: 10.1186/S12957-022-02772-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ahmadipour Y, Rauschenbach L, Santos A, et al. Preoperative and early postoperative seizures in patients with glioblastoma—two sides of the same coin? Neurooncol Adv. 2021;3(1):1–9. doi: 10.1093/NOAJNL/VDAA158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Dührsen L, Sauvigny T, Ricklefs FL, et al. Seizures as presenting symptom in patients with glioblastoma. Epilepsia. 2019;60(1):149–154. doi: 10.1111/EPI.14615 [DOI] [PubMed] [Google Scholar]
- 69.Marku M, Rasmussen BK, Belmonte F, et al. Prediagnosis epilepsy and survival in patients with glioma: a nationwide population-based cohort study from 2009 to 2018. J Neurol. 2022;269(2):861–872. doi: 10.1007/S00415-021-10668-6/TABLES/3 [DOI] [PubMed] [Google Scholar]
- 70.Rudà R, Horbinski C, van den Bent M, et al. IDH inhibition in gliomas: from preclinical models to clinical trials. Nat Rev Neurol. 2024;20(7):395–407. doi: 10.1038/s41582-024-00967-7 [DOI] [PubMed] [Google Scholar]
- 71.Songtao Q, Lei Y, Si G, et al. IDH mutations predict longer survival and response to temozolomide in secondary glioblastoma. Cancer Sci. 2012;103(2):269–273. doi: 10.1111/J.1349-7006.2011.02134.X [DOI] [PubMed] [Google Scholar]
- 72.Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(8):1231. doi: 10.1093/NEUONC/NOAB106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Walter FM, Penfold C, Joannides A, et al. Missed opportunities for diagnosing brain tumours in primary care: a qualitative study of patient experiences. Br J Gen Pract. 2019;69(681):E224–E234. doi: 10.3399/BJGP19X701861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mastall M, Wolpert F, Gramatzki D, et al. Survival of brain tumour patients with epilepsy. Brain. 2021;144(11):3322–3327. doi: 10.1093/BRAIN/AWAB188 [DOI] [PubMed] [Google Scholar]
- 75.Flanigan PM, Jahangiri A, Truong A, et al. Improved survival with decreased Wait Time to surgery in glioblastoma patients presenting with Seizure Ruby Kuang, MS §. Neurosurgery. doi: 10.1093/neuros/nyx084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bette S, Barz M, Nham HL, et al. Image analysis reveals microstructural and volumetric differences in glioblastoma patients with and without preoperative seizures. Cancers (Basel). 2020;12(4). doi: 10.3390/CANCERS12040994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Lee JW, Wen PY, Hurwitz S, et al. Morphological characteristics of brain tumors causing seizures. Arch Neurol. 2010;67(3):336–342. doi: 10.1001/ARCHNEUROL.2010.2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chaichana KL, Parker SL, Olivi A, et al. Long-term seizure outcomes in adult patients undergoing primary resection of malignant brain astrocytomas: clinical article. J Neurosurg. 2009;111(2):282–292. doi: 10.3171/2009.2.JNS081132 [DOI] [PubMed] [Google Scholar]
- 79.Henker C, Kriesen T, Scherer M, et al. Association between tumor compartment volumes, the incidence of pretreatment seizures, and statin-mediated protective effects in glioblastoma. Neurosurgery. 2019;85(4):E722–E729. doi: 10.1093/NEUROS/NYZ079 [DOI] [PubMed] [Google Scholar]
- 80.Sanai N, Polley MY, Mw M, et al. An extent of resection threshold for newly diagnosed glioblastomas: clinical article. J Neurosurg. 2011;115(1):3–8. doi: 10.3171/2011.2.JNS10998 [DOI] [PubMed] [Google Scholar]
- 81.Skardelly M, Brendle E, Noell S, et al. Predictors of preoperative and early postoperative seizures in patients with intra-axial primary and metastatic brain tumors: a retrospective observational single center study. Ann Neurol. 2015;78(6):917–928. doi: 10.1002/ANA.24522 [DOI] [PubMed] [Google Scholar]
- 82.Shen S, Bai Y, Zhang B, et al. Correlation of preoperative seizures with a wide range of tumor molecular markers in gliomas: an analysis of 442 glioma patients from China. Epilepsy Res. 2020:166. doi: 10.1016/J.EPLEPSYRES.2020.106430 [DOI] [PubMed] [Google Scholar]
- 83.Chaichana KL, Halthore AN, Parker SL, et al. Factors involved in maintaining prolonged functional independence following supratentorial glioblastoma resection. J Neurosurg. 2011;114(3):604. doi: 10.3171/2010.4.JNS091340 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Climans SA, Brandes AA, Cairncross JG, et al. Temozolomide and seizure outcomes in a randomized clinical trial of elderly glioblastoma patients. J Neurooncol. 2020;149(1):65–71. doi: 10.1007/S11060-020-03573-X/TABLES/3 [DOI] [PubMed] [Google Scholar]
- 85.Shin JY, Kizilbash SH, Robinson S, et al. Incidence, characteristics, and implications of seizures in patients with glioblastoma. Am J Hosp Palliat Care. 2017;34(7):650–653. doi: 10.1177/1049909116647405 [DOI] [PubMed] [Google Scholar]
- 86.Rabin EE, Huang J, Kim M, et al. Age-stratified comorbid and pharmacologic analysis of patients with glioblastoma. Brain Behav Immun Health. 2024;38:100753. doi: 10.1016/J.BBIH.2024.100753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Seidenberg M, Pulsipher DT, Hermann B. Association of epilepsy and comorbid conditions. Future Neurol. 2009;4(5):663. doi: 10.2217/FNL.09.32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. doi: 10.1056/NEJMOA043330/SUPPL_FILE/987SA1.PDF [DOI] [PubMed] [Google Scholar]
- 89.Bobustuc GC, Baker CH, Limaye A, et al. Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro Oncol. 2010;12(9):917–927. doi: 10.1093/NEUONC/NOQ044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Scicchitano BM, Sorrentino S, Proietti G, et al. Levetiracetam enhances the temozolomide effect on glioblastoma stem cell proliferation and apoptosis. Cancer Cell Int. 2018;18(1). doi: 10.1186/S12935-018-0626-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Marutani A, Nakamura M, Nishimura F, et al. Tumor-inhibition effect of levetiracetam in combination with temozolomide in glioblastoma cells. Neurochem J. 2017;11(1):43–49. doi: 10.1134/S1819712416040073/METRICS [DOI] [Google Scholar]
- 92.Roh TH, Moon JH, Park HH, et al. Association between survival and levetiracetam use in glioblastoma patients treated with temozolomide chemoradiotherapy. Sci Rep. 2020;10(1):1–7. doi: 10.1038/s41598-020-67697-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kim YH, Kim T, Joo JD, et al. Survival benefit of levetiracetam in patients treated with concomitant chemoradiotherapy and adjuvant chemotherapy with temozolomide for glioblastoma multiforme. Cancer. 2015;121(17):2926–2932. doi: 10.1002/CNCR.29439 [DOI] [PubMed] [Google Scholar]
- 94.Ryu JY, Min KL, Chang MJ. Effect of anti-epileptic drugs on the survival of patients with glioblastoma multiforme: a retrospective, single-center study. PLOS ONE. 2019;14(12). doi: 10.1371/JOURNAL.PONE.0225599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Pallud J, Huberfeld G, Dezamis E, et al. Effect of levetiracetam use duration on overall survival of isocitrate dehydrogenase wild-type glioblastoma in adults: an observational study. Neurology. 2022;98(2):E125–E140. doi: 10.1212/WNL.0000000000013005 [DOI] [PubMed] [Google Scholar]
- 96.Jabbarli R, Ahmadipour Y, Rauschenbach L, et al. How about levetiracetam in glioblastoma? An institutional experience and meta-analysis. Cancers (Basel). 2021;13(15). doi: 10.3390/CANCERS13153770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hwang K, Kim J, Kang SG, et al. Levetiracetam as a sensitizer of concurrent chemoradiotherapy in newly diagnosed glioblastoma: an open-label phase 2 study. Cancer Med. 2022;11:371–379. doi: 10.1002/cam4.4454 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chen JS, Clarke R, Haddad AF, et al. The effect of levetiracetam treatment on survival in patients with glioblastoma: a systematic review and meta-analysis. J Neurooncol. 2022;156(2):257–267. doi: 10.1007/S11060-021-03940-2 [DOI] [PubMed] [Google Scholar]
- 99.Knudsen-Baas KM, Storstein AM, Zarabla A, et al. Antiseizure medication in patients with glioblastoma- a collaborative cohort study. Seizure. 2021;87:107–113. doi: 10.1016/J.SEIZURE.2021.03.012 [DOI] [PubMed] [Google Scholar]
- 100.Bianconi A, Koumantakis E, Gatto A, et al. Effects of Levetiracetam and lacosamide on survival and seizure control in idh-wild type glioblastoma during temozolomide plus radiation adjuvant therapy. Brain Spine. 2024;4:2772–5294. doi: 10.1016/J.BAS.2023.102732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sun M, Huang N, Tao Y, et al. The efficacy of temozolomide combined with levetiracetam for glioblastoma (GBM) after surgery: a study protocol for a double-blinded and randomized controlled trial. Trials. 2022;23(1). doi: 10.1186/S13063-022-06168-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Phiel CJ, Zhang F, Huang EY, et al. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276(39):36734–36741. doi: 10.1074/JBC.M101287200 [DOI] [PubMed] [Google Scholar]
- 103.Knüpfer MM, Hernáiz-Driever P, Poppenborg H, et al. Valproic acid inhibits proliferation and changes expression of CD44 and CD56 of malignant glioma cells in vitro. Anticancer Res. 1998. [cited 2024 Aug 16];18(5A):3585–3589. Available from: https://europepmc.org/article/med/9858943 [PubMed] [Google Scholar]
- 104.Yang ZY, Wang XH. Valproic acid inhibits glioma and its mechanisms. J Healthc Eng. 2022;2022. doi: 10.1155/2022/4985781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Osuka S, Takano S, Watanabe S, et al. Valproic acid inhibits angiogenesis in vitro and glioma angiogenesis in vivo in the brain. Neurol Med Chir (Tokyo). 2012;52(4):186–193. doi: 10.2176/NMC.52.186 [DOI] [PubMed] [Google Scholar]
- 106.Tsai HC, Wei KC, Chen PY, et al. Valproic acid enhanced temozolomide-induced anticancer activity in human glioma through the p53-puma apoptosis pathway. Front Oncol. 2021:11. doi: 10.3389/FONC.2021.722754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Chen CH, Chang YJ, Msb K, et al. Enhancement of temozolomide-induced apoptosis by valproic acid in human glioma cell lines through redox regulation. J Mol Med (Berl). 2011;89(3):303–315. doi: 10.1007/S00109-010-0707-1 [DOI] [PubMed] [Google Scholar]
- 108.Ryu CH, Yoon WS, Park KY, et al. Valproic acid downregulates the expression of MGMT and sensitizes temozolomide-resistant glioma cells. J Biomed Biotechnol. 2012;2012. doi: 10.1155/2012/987495 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Camphausen K, Cerna D, Scott T, et al. Enhancement of in vitro and in vivo tumor cell radiosensitivity by valproic acid. Int J Cancer. 2005;114(3):380–386. doi: 10.1002/IJC.20774 [DOI] [PubMed] [Google Scholar]
- 110.Thotala D, Karvas RM, Engelbach JA, et al. Valproic acid enhances the efficacy of radiation therapy by protecting normal hippocampal neurons and sensitizing malignant glioblastoma cells. Oncotarget. 2015;6(33):35004. doi: 10.18632/ONCOTARGET.5253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sears TK, Horbinski CM, Woolard KD. IDH1 mutant glioma is preferentially sensitive to the HDAC inhibitor panobinostat. J Neurooncol. 2021;154(2):159–170. doi: 10.1007/S11060-021-03829-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Watanabe S, Kuwabara Y, Suehiro S, et al. Valproic acid reduces hair loss and improves survival in patients receiving temozolomide-based radiation therapy for high-grade glioma. Eur J Clin Pharmacol. 2017;73(3):357–363. doi: 10.1007/S00228-016-2167-1/FIGURES/3 [DOI] [PubMed] [Google Scholar]
- 113.Barker CA, Bishop AJ, Chang M, et al. Valproic acid use during radiation therapy for glioblastoma associated with improved survival. Int J Radiat Oncol Biol Phys. 2013;86(3):504. doi: 10.1016/J.IJROBP.2013.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Yuan Y, Xiang W, Qing M, et al. Survival analysis for valproic acid use in adult glioblastoma multiforme: a meta-analysis of individual patient data and a systematic review. Seizure. 2014;23(10):830–835. doi: 10.1016/J.SEIZURE.2014.06.015 [DOI] [PubMed] [Google Scholar]
- 115.Sullivan JK, Fahey PP, Agho KE, et al. Valproic acid as a radio-sensitizer in glioma: a systematic review and meta-analysis. Neurooncol Pract. 2023;10(1):13–23. doi: 10.1093/NOP/NPAC078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kuo YJ, Yang YH, Lee IY, et al. Effect of valproic acid on overall survival in patients with high-grade gliomas undergoing temozolomide a nationwide population-based cohort study in Taiwan. Published online 2020. doi: 10.1097/MD.0000000000021147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Happold C, Gorlia T, Chinot O, et al. Does valproic acid or levetiracetam improve survival in glioblastoma? A pooled analysis of prospective clinical trials in newly diagnosed glioblastoma. J Clin Oncol. 2016;34(7):731–739. doi: 10.1200/JCO.2015.63.6563 [DOI] [PMC free article] [PubMed] [Google Scholar]; • Population-wide study which demonstrated increased survival of high-grade glioma with valproic acid, although patients with survival benefit were younger and molecular markers were not considered.
- 118.Jenkinson MD, Bulbeck H, Rooney A, et al. P18.39. A SEIZURE PROPHYLAXIS in GLIOMA SURGERY (SPRING): A MULTI-CENTRE, UNBLINDED, RANDOMISED TRIAL. Neuro Oncol. 2024;26(Supplement_5):v106–v106. doi: 10.1093/NEUONC/NOAE144.355 [DOI] [Google Scholar]
- 119.Drumm M, Wang W, Sears TK, et al. Postoperative risk of IDH mutant glioma-associated seizures and their potential management with IDH mutant inhibitors. J Clin Invest. [cited 2023 Apr 27]. doi: 10.1172/JCI168035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Vo AH, Ambady P, Spencer D. The IDH1 inhibitor ivosidenib improved seizures in a patient with drug-resistant epilepsy from IDH1 mutant oligodendroglioma. doi: 10.1016/j.ebr.2022.100526 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Galanis E, Keith Anderson S, Miller CR, et al. Phase I/II trial of vorinostat combined with temozolomide and radiation therapy for newly diagnosed glioblastoma: results of alliance N0874/ABTC 02. Neuro Oncol. 2018;20(4):546–556. doi: 10.1093/NEUONC/NOX161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Iwamoto FM, Lamborn KR, Kuhn JG, et al. A phase I/II trial of the histone deacetylase inhibitor romidepsin for adults with recurrent malignant glioma: north American brain tumor consortium study 03–03. Neuro Oncol. 2011;13(5):509–516. doi: 10.1093/NEUONC/NOR017 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Review on the anti-glioma properties of valproic acid.
- 123.Han W, Guan W. Valproic acid: a promising therapeutic agent in glioma treatment. Front Oncol. 2021;11:687362. doi: 10.3389/FONC.2021.687362/BIBTEX [DOI] [PMC free article] [PubMed] [Google Scholar]
