Skip to main content

MINI REVIEW article

Front. Pharmacol., 06 October 2022
Sec. Neuropharmacology
This article is part of the Research Topic Experimental Models of Epilepsy and Related Comorbidities, Volume II View all 7 articles

Epilepsy treatment in neuro-oncology: A rationale for drug choice in common clinical scenarios

  • 1Department of Neurology, University Hospital Complex of Cartagena, Murcia, Spain
  • 2Department of Cell Biology and Histology, School of Medicine, Regional Campus of International Excellence, “Campus Mare Nostrum”, IMIB-Arrixaca, University of Murcia, Murcia, Spain
  • 3Epilepsy Program, Department of Neurology, Ruber International Hospital, Madrid, Spain
  • 4Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
  • 5Department of Radiology and Physical Medicine, School of Medicine, Regional Campus of International Excellence, “Campus Mare Nostrum”, IMIB-Arrixaca, University of Murcia, Murcia, Spain

Epilepsy represents a challenge in the management of patients with brain tumors. Epileptic seizures are one of the most frequent comorbidities in neuro-oncology and may be the debut symptom of a brain tumor or a complication during its evolution. Epileptogenic mechanisms of brain tumors are not yet fully elucidated, although new factors related to the underlying pathophysiological process with possible treatment implications have been described. In recent years, the development of new anti-seizure medications (ASM), with better pharmacokinetic profiles and fewer side effects, has become a paradigm shift in many clinical scenarios in neuro-oncology, being able, for instance, to adapt epilepsy treatment to specific features of each patient. This is crucial in several situations, such as patients with cognitive/psychiatric comorbidity, pregnancy, or advanced age, among others. In this narrative review, we provide a rationale for decision-making in ASM choice for neuro-oncologic patients, highlighting the strengths and weaknesses of each drug. In addition, according to current literature evidence, we try to answer some of the most frequent questions that arise in daily clinical practice in patients with epilepsy related to brain tumors, such as, which patients are the best candidates for ASM and when to start it, what is the best treatment option for each patient, and what are the major pitfalls to be aware of during follow-up.

1 Introduction

Epileptic seizures are one of the most frequent comorbidities in neuro-oncology and can be either the initial symptom of a brain tumor or a complication during its evolution. Epilepsy is more frequent in primary tumors than in brain metastases (Glantz et al., 2000), although the latter represent the most frequent intracranial tumor (Sánchez-Villalobos et al., 2021b). The prevalence of epilepsy also varies among primary neoplasms according to tumor type and grade, being diffuse low-grade gliomas one of the most highly epileptogenic (Glantz et al., 2000; Pallud et al., 2014).

Currently, epilepsy is a major risk factor for long-term disability in patients with brain tumor-related epilepsy (BTRE) (Maschio, 2012). This is not only due to the negative impact of seizures on quality of life (Rudà et al., 2012), but also to the morbidity associated with both somatic and neuropsychiatric side effects of antiseizure medications (ASM) (Kanner, 2016a). To date, the evidence regarding the use of ASM in BTRE patients is limited. It is overall recommended not to use those drugs with a greater enzyme-inducing effect, given the possibility of modifying the metabolism of antineoplastic drugs. The large availability of ASMs increases both the complexity of drug choice and the possibilities for tailoring treatments according to pharmacokinetics, drug-to-drug interactions, or comorbidities profile, among other factors, such as neoplasm type or genetic profile (Beltrán-Corbellini et al., 2022).

In this narrative review, we provide an overview of ASM in neuro-oncology to help with decision-making, focusing on glial tumors and highlighting the strengths and weaknesses of each ASM. In addition, according to the current evidence, this paper assesses some of the most relevant questions that arise in daily clinical practice in patients with BTRE, such as: i) which patients are the best candidates for ASM prescription; ii) when to initiate ASM; or, iii) which is the best treatment option for each patient concerning their comorbidities or clinical profiles.

2 Brief summary on molecular factors in epileptogenesis of brain tumors

Epileptogenesis of brain tumors is influenced by many factors, including tumor location, histological characteristics of the neoplasm, changes in neurotransmitter homeostasis and the peritumoral environment, changes in the integrity of the blood-brain barrier, as well as genetic factors (Ertürk Çetin et al., 2017). To date, several biological and molecular factors have been described that could be involved in the epileptogenesis of brain tumors. Some of them are listed below: with respect to glutamate, high concentrations found in the peritumoral environment would contribute to an increased risk of seizure development and recurrence (Rosati et al., 2010; Goldstein and Feyissa, 2018; Neal et al., 2016). In gliomas, this increase in synaptic concentrations is due to changes in glial membrane transporter systems (De Groot et al., 2011). In addition, glutamergic stimulation of N-methyl-d-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors can activate the intracellular signaling pathways of mammalian target of rapamycin (mTOR), AKT and mitogen-activated protein kinase (MAPK), contributing to both cell growth and epilepsy (De Groot et al., 2011; Englot et al., 2016a). GABAergic signaling is also implicated in both tumor growth and paradoxical excitatory effects mediated by alterations in neuronal and tumor cell chloride ion homeostasis related to cotransporter changes (Pallud et al., 2014). Finally, another factor studied focuses on the isocitrate dehydrogenase 1 (IDH1) enzyme. IDH catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate, while in its mutated form, it reduces α-ketoglutarate to D-2-hydroxyglutarate (D2HG) (Turkalp et al., 2014). The D2HG product of IDH1mut can increase neuronal activity by mimicking glutamate activity at the NMDA receptor, and IDH1mut gliomas are more likely to cause seizures in patients (Chen et al., 2016). These represent only some of the molecular factors related to epileptogenesis in brain tumors, other factors such as O-6-methylguanine DNA methyltransferase (MGMT), MMP-9, BDNF, p53 and adenosine kinase (ADK) have also been proposed (Goldstein and Feyissa, 2018).

3 Treatment indication in brain tumor-related seizures: When to start and stop antiseizure medication

First, in patients with brain tumors who present with a first seizure, even in the absence of pathological findings on electroencephalogram (EEG) or a second seizure, ASM should be initiated, due to the high risk of recurrence (Chen et al., 2018). Second, there is currently sufficient evidence to discourage treatment with ASM in patients with brain tumors who did not present any seizures (Glantz et al., 2000; Englot et al., 2016a; Chen et al., 2018).

Third, regarding the perioperative use of ASM in patients with brain tumors, a recent Cochrane systematic review did not find evidence of the effectiveness of ASMs (Greenhalgh et al., 2020). Nevertheless, the addition of prophylactic ASM is perioperatively recommended in patients with brain tumors undergoing craniotomy. This treatment should be withdrawn 1 week after surgery (Kuijlen et al., 1996; Glantz et al., 2000; Iuchi et al., 2015; Englot et al., 2016a; Pourzitaki et al., 2016).

Finally, there is no current evidence-based recommendation or consensus on the duration of treatment for epilepsy-related to brain tumors (Chen et al., 2018). Among the factors to be considered in this clinical scenario, we suggest: i) optimal seizure control; ii) complete resection (or not) of the tumor; iii) EEG findings; iv) social and working particularities; v) individualized decision in agreement with patient and caregiver.

4 Antiseizure medication for brain tumor-related epilepsy

Currently, the availability of studies evaluating ASM efficacy in patients with BTRE is scarce. Nevertheless, given that epilepsy in these patients is thought to be secondary to a focal brain lesion, usually, the treatment scheme is similar to that of focal-onset epilepsies (Chen et al., 2018). Although the approach to seizures in BTRE patients is multidisciplinary and involves medical, radiotherapeutic, and surgical treatment, in his review we will focus on the use of ASMs. Similarly, although the main target of this article is the control of epilepsy in patients with glial tumors, given that many of the aspects described here are extensive to other lesions, we have considered it necessary to include a comparative table with the main clinical and epidemiological characteristics of the main intracranial lesions (Table 1). For each drug, we will describe the main aspects related to the mechanism of action, pharmacokinetics, main adverse effects, as well as the evidence on the drug in BTRE (Table 2 and Figure 1).

TABLE 1
www.frontiersin.org

TABLE 1. Main characteristics of brain tumors and their relationship with epilepsy.

TABLE 2
www.frontiersin.org

TABLE 2. Commonly used antiseizure medications in patients with brain tumor related epilepsy.

FIGURE 1
www.frontiersin.org

FIGURE 1. Scheme of the mechanism of action of antiseizure medications. *ASM with more than one proposed mechanism of action. Modified from (Löscher and Klein, 2021). Abbreviations: AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid; GABA, gamma-aminobutyric acid; GLU, glutamate; NMDA, N-methyl-d-aspartate.

4.1 Synaptic vesicles protein 2A binders

4.1.1 Levetiracetam

Levetiracetam is an (S)-enantiomer of the ethyl analog of piracetam (Wright et al., 2013). Although the precise mechanism is unknown, in animal models it has been shown to bind to the synaptic vesicle protein 2a (SV2a) (Lynch et al., 2004), an integral transmembrane glycoprotein ubiquitously expressed in all synaptic terminals (Contreras-García et al., 2021).

In pharmacokinetics, most (66%) of levetiracetam is eliminated through the kidneys (Hovinga, 2001). No posology adjustment is needed for patients with hepatic impairment (Wright et al., 2013). Other advantages include rapid and almost complete absorption via oral (96%), low plasma protein binding (<10%), oral and intravenous formulation, and a safety profile with a high therapeutic index and low drug-to-drug interactions (Klitgaard et al., 1998; Wright et al., 2013).

Levetiracetam is frequently prescribed in BTRE patients, being one of the most widely used first-line ASM (Sánchez-Villalobos et al., 2021a). Numerous studies have shown the efficacy of levetiracetam in BTRE patients both in monotherapy (Dinapoli et al., 2009; Merrell et al., 2010; Rosati et al., 2010; Usery et al., 2010; Maschio et al., 2011b; De Groot et al., 2011; Bähr et al., 2012; Rossetti et al., 2014; Berntsson et al., 2018; Cardona et al., 2018; Casas Parera et al., 2019; Kerkhof et al., 2019; Ius et al., 2020; Solomons et al., 2020) and in polytherapy (Wagner et al., 2003; Maschio et al., 2006; Newton et al., 2006; Van Breemen et al., 2009; Haggiagi and Avila, 2019; Chonan et al., 2020; Rudà et al., 2020). In a recent systematic review (Bruin et al., 2021), patients with seizures secondary to grade II-IV gliomas treated with levetiracetam monotherapy had a 6-months seizure freedom rate of 39–96%, with a 6-months failure rate due to adverse effects and ineffectiveness of 1% and 10%, respectively.

As the main side effects, levetiracetam exhibits some relevant downsides, including psychiatric iatrogenic symptoms (7–25%) (Kanner and Bicchi, 2022), such as depression, anxiety, psychosis and behavioral disturbances (Dinkelacker et al., 2003; Dannaram et al., 2012; Lin et al., 2012; Chen et al., 2016; Thelengana et al., 2019). Moreover, patients in treatment with levetiracetam experience more frequent adverse psychiatric effects than those with the other ASMs (Weintraub et al., 2007). In addition, patients with frontal lobe tumors may be at increased risk of neuropsychiatric adverse effects with levetiracetam (Bedetti et al., 2017). Add-on treatment with pyridoxine for the control of levetiracetam-induced behavioral adverse effects might be considered in some patients (Marino et al., 2018; Dreischmeier et al., 2021).

Finally, epigenetic silencing of the MGMT enzyme by levetiracetam could lead to an “antitumor” effect, by increasing the temozolomide efficacy (Bobustuc et al., 2010; Kim et al., 2015). Moreover, a recent study suggests that the use of levetiracetam throughout standard chemoradiation protocol possibly improves the overall survival of patients with isocitrate dehydrogenase (IDH) wild-type glioblastoma (Pallud et al., 2021). However, other previous studies did not show any improvement in the survival of levetiracetam in patients with newly diagnosed glioblastoma (Happold et al., 2016). Thus, further studies are warranted in the future to clarify the potential survival improvement effect.

In summary, levetiracetam has been shown to be a safe and effective drug in BTRE patients, although neuropsychiatric effects should be monitored.

4.1.2 Brivaracetam

Brivaracetam is a selective, reversible, high-affinity ligand of SV2A (15–30 fold higher than levetiracetam) (de Biase et al., 2020). Pharmacokinetically, brivaracetam has the ability to rapidly cross the blood-brain barrier due to its lipophilicity, which is similar to that of benzodiazepines and higher than levetiracetam (Niespodziany et al., 2017). In addition, brivaracetam and levetiracetam, are useful for the treatment of status epilepticus (Santamarina et al., 2019), which makes them both an interesting option in emergency situations. Brivaracetam is extensively metabolized in the liver. Thus, its dose needs to be reduced in patients with liver damage regardless of the Child-Pugh score (de Biase et al., 2020). In contrast, brivaracetam does not induce or inhibit CYP enzymes or the known drug transport system, except for CYP2C19 (weakly inhibited in vitro studies). Thus, it has a low potential for clinically relevant drug interactions (de Biase et al., 2020).

To date, Maschio et al. have published the only retrospective study of BTRE-patients treated with brivaracetam as add-on therapy (n = 33). In that study, patients had a high responder rate (78.8%) with a mean follow-up of 10 months. The main cause of drug discontinuation was, again, psychiatric adverse effects (9%) (Maschio et al., 2020a). Although no specific clinical trials comparing psychiatric adverse effects between levetiracetam and brivaracetam are available to date, some studies in non-oncological population show slightly fewer psychiatric adverse effects with brivaracetam (Feyissa, 2019; Villanueva et al., 2019).

Finally, despite being a novel drug, brivaracetam could be considered as an option to be evaluated in BTRE patients, although further studies are needed to unveil both efficacy and tolerability in this population.

4.2 Sodium channel blockers

Sodium channel blockers (SCBs) are one of the main families of ASM. We will divide them into different groups: lacosamide, dibenzazepines and lamotrigine.

4.2.1 Lacosamide

Lacosamide is an ASM that selectively increases the slow inactivation of voltage-gated sodium channels, stabilizing the voltage-gated neuronal membranes. In addition, lacosamide appears to interact with collapsing-response mediator protein 2 (CRMP2), thereby enhancing neuronal plasticity (Kellinghaus, 2009). The main pharmacokinetic strengths of lacosamide are low protein binding (less than 15%), no inhibition or induction of several of the hepatic microsomal isoenzymes of importance (CYP2C19 and CYP3A4) to a clinically relevant degree and very low potential for drug-to-drug interactions (Sánchez-Villalobos et al., 2018). Another strength of lacosamide is the possibility of intravenous use in emergency situations requiring rapid uptitration, such as status epilepticus (Strzelczyk et al., 2017). Currently, several studies of lacosamide in polytherapy in BTRE patients have been published (Maschio et al., 2011a; Saria et al., 2013; Villanueva et al., 2016; Maschio et al., 2017b; Rudà et al., 2018; Rudà et al., 2020). The VIBES study, a prospective study (n = 93) that analyzed the efficacy and tolerability of lacosamide as add-on therapy in patients diagnosed of BTRE secondary to low-grade glioma (WHO grade I-II), showed at 6 months a ≥50% reduction in seizure frequency from baseline in 76.7% of patients and being 34.9% seizure-free. 4.3% of patients had drug effects leading to discontinuation (Rudà et al., 2020). Recently, the first retrospective study (n = 132) analyzing the efficacy and tolerability of lacosamide in monotherapy in BTRE has been published, showing absence of seizures in 64.4% of patients after 3 months and 55% after 6 months, with a low dropout rate (1.5%) (Mo et al., 2022).

Regarding adverse effects, these are usually mild and dose-related, sometimes more evident after the morning peak dose, being dizziness and drowsiness the most frequent ones (Mo et al., 2022). On the contrary, it is contraindicated in patients with second and third-degree atrioventricular block. Lacosamide also has proven evidence in treating neuropathic and inflammatory pain in various animal models and observational studies in humans (Stöhr et al., 2006; Alcántara-Montero and Sánchez-Carnerero, 2016; Sánchez-Villalobos et al., 2018), while in the psychiatric sphere, it behaves as a fairly neutral drug (Kanner, 2016a). Finally, in vitro antineoplastic effect of lacosamide and brivaracetam in human glioma cells was recently reported (Rizzo et al., 2017).

4.2.2 Dibenzazepines

There are three available different drugs in the dibenzazepine family, from the oldest to the most recent: carbamazepine, oxcarbazepine and eslicarbazepine acetate.

According to their pharmacodynamics, carbamazepine and oxcarbazepine act by blocking the fast inactivation state of gated sodium channels, while eslicarbazepine acetate blocks sodium channel’s slow inactivation. Regarding pharmacokinetics, the main issue is enzymatic induction, which is less pronounced for oxcarbazepine and eslicarbazepine acetate than for carbamazepine. However, carbamazepine shows lower risk of hyponatremia, and larger antiseizure effectiveness in comparative studies (Aledo-Serrano and Gil-Nagel, 2020). There is previous experience in BTRE-patients treated with carbamazepine (Warnke et al., 1997; Zaatreh et al., 2002; Zaatreh et al., 2003; Wick et al., 2005), oxcarbazepine (Maschio et al., 2009; 2012a) and more recently and to a more limited extent with eslicarbazepine acetate (Leslie et al., 2020; Zoccarato et al., 2021). A remarkable aspect of these drugs is that they can have a positive effect in the psychiatric sphere, for example, as mood stabilizers (Kanner, 2016a). Since carbamazepine, as well as phenytoin, are major enzyme inducers, they would not be recommended as first-line treatment in BTRE-patients.

4.2.3 Lamotrigine

Lamotrigine is a first-line ASM for the treatment of focal epilepsy, without enzyme induction features (Perucca and Tomson, 2011). Among its main disadvantages, the need for slow titration and the risk of allergic reactions, mainly skin-related but potentially severe, are of notice, along with the interaction with valproate, which may influence a rigorous dose monitoring (Bruin et al., 2021). This may make lamotrigine an unsuitable starting option in BTRE-patients who needs rapid treatment. With good pharmacokinetics and adverse effects profile, lamotrigine might be a good option in other clinical scenarios.

4.3 Valproic acid

The mechanisms of action of valproic acid are not yet fully understood, but its effect on the synthesis and release of γ-aminobutyric acid (GABA) is important, as it increases the effect of GABA in certain brain regions. In addition, the effect on the N-methyl-d-aspartate (NMDA) receptor appears to play an important role in its anti-seizure effect. Pharmacokinetically, the oral bioavailability rate of valproate is close to 100% and approximately 85–95% of the absorbed valproate dose is bound to plasma proteins. In patients with renal insufficiency, chronic hepatic insufficiency or elderly patients, the protein-bound portion is reduced (Baumgartner and Elger, 2020). One of the main problems with valproate is that it inhibits multiple components of CYP system. This might lead to decreased metabolism of some chemotherapeutic agents, increasing their toxicity. Regarding the current evidence, valproate is one of the most historically prescribed ASMs in epilepsy. It is a broad-spectrum ASM that has been used for decades. It is effective in the treatment of focal epilepsies as well as in all types of generalized epilepsy. Similarly, there is extensive experience with the use of valproate in BTRE-patients, both in monotherapy and in polytherapy (Zaatreh et al., 2002; Zaatreh et al., 2003; Wick et al., 2005; Van Breemen et al., 2009; Simó et al., 2012; You et al., 2012; Kerkhof et al., 2013; Yuan et al., 2013).

The most common side effects include weight gain, gastrointestinal complaints, hair loss, hirsutism, and tremor. However, one of the most relevant is thrombocytopenia (12–18% of treated individuals), with advanced age, female sex and high doses of the drug as main risk factors. In addition, the administration of valproate combined with nitrosoureas, etoposide and cisplatin increases bone marrow toxicity, as well as the combination with temozolomide is associated with an increased risk of thrombocytopenia and neutropenia (Bourg et al., 2001; Simó et al., 2012; Bruna et al., 2013).

Finally, it is noteworthy that valproate is associated with increased survival in several observational studies, when administered during chemoradiation therapy in patients with glioblastoma (Weller et al., 2011; Kerkhof et al., 2013; Krauze et al., 2015). Proposed mechanisms would involve increased bioavailability of temozolamide or the histone deacetylase inhibitory activity of valproate, with subsequent sensitization of glioblastoma cells to chemoradiation (Weller et al., 2011; Krauze et al., 2015). However, recently Happold et al. (2016) performed a pooled analysis of the survival association of ASM use at the initiation of chemoradiotherapy with temozolomide (n = 1.869 within four randomized clinical trials) in newly diagnosed glioblastoma, with no survival improvement among patients treated with valproate (and/or levetiracetam) (Happold et al., 2016).

4.4 Others

4.4.1 Calcium channel α2δ-subunit blockers

Pregabalin and gabapentin are α2δ-subunit of calcium channel blockers. Although these drugs were initially used for the treatment of seizures, they are now more commonly used for the treatment of neuropathic pain. Nevertheless, pregabalin could represent a valid alternative as add-on therapy in BTRE patients, especially in those with comorbidities such as neuropathic pain or anxiety (Maschio et al., 2012b; Rossetti et al., 2014).

4.4.2 Perampanel

Perampanel is a highly selective, noncompetitive, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)-type glutamate receptor antagonist. Although it is a relatively novel ASM, some studies have already demonstrated its efficacy as an add-on therapy in BTRE patients (Vecht et al., 2017; Izumoto et al., 2018; Maschio et al., 2019; Maschio et al., 2020b; Chonan et al., 2020). Perampanel presents weak enzyme induction at high doses and require a single daily dose. Additionally, some studies show a positive impact on sleep architecture, as well as relevant side effects in the neuropsychiatric sphere in a subgroup of patients (Kanner, 2016a; Rocamora et al., 2020). Finally, recently some in-vitro studies have shown a pro-apoptotic effect of perampanel in human glioblastoma cell lines when used alone, possibly due to increased GluR2/3 expression, as well as a possible synergistic effect when used in combination with temozolamide (Salmaggi et al., 2021).

4.4.3 Topiramate and zonisamide

Topiramate and zonisamide bind to sodium channels and voltage-sensitive calcium channels. Both have been previously used in BTRE-patients and do not present clinically significant enzyme induction features, being an alternative in this patient population (Maschio et al., 2008, 2017a; Lu et al., 2009). Zonisamide has among its advantages a single daily dosage and minimal drug-drug interaction. Among down-sides for both drugs, intravenous formulation is not available, and they show side effects with potential negative impact on cognition and weight loss (Goldstein and Feyissa, 2018). Finally, these drugs are not recommended in patients with gliobastoma and/or high-grade astrocytoma, given their potential side effect with metabolic acidosis and therefore interaction with temozolamide (Grupo Español de Investigación en Neurooncología, 2021).

5 Considerations according to particular situations

5.1 Elderly patients

Incidence rate of glioblastoma among elderly patients (aged 70 years or older) is 17.5 per 100,000 person-years, representing a relative risk of 3–4 times compared to young adults (Minniti et al., 2019). Therefore, it is interesting to address some of the particularities of ASM in this population. Aging is accompanied by several physiological changes, which affect both the ASM pharmacokinetic and pharmacodynamic characteristics. On the one hand, since renal clearance decreases with aging, the doses of ASMs should be adjusted with renal function. On the other hand, since liver function progressively decreases with aging, the consequent reduction in serum albumin could lead to an elevation of the free fraction of some ASM, potentially increasing the risk of adverse effects. Therefore, liver function should be closely monitored in the elderly patient treated with ASMs (Italiano and Perucca, 2013). Classical ASMs such as phenytoin, carbamazepine, or phenobarbital with a higher enzyme induction profile could reduce plasma concentrations not only of antineoplastic drugs, but also with other drugs commonly taken by elderly patients, such as anticoagulants, antidepressants or antimicrobials (Seo et al., 2020). Likewise, valproate, with an enzyme inhibitor effect, could increase the serum concentrations of some of them, or in the case of some antineoplastic drugs such as temozolamide, increase the hematological toxicity (Simó et al., 2012). And finally, the impact of the ASM on cognition should also be taken into account with phenytoin, topiramate or zonisamide, being some of the ASM that can produce cognition impairment in elderly patients (Seo et al., 2020).

5.2 Epilepsy and pregnancy

The possibility of gestation in a woman with BTRE adds a new dimension to the challenge of choosing and subsequently managing ASMs. The risks associated with the use of ASMs during pregnancy are a major concern for all women of childbearing age with epilepsy. Indeed, both the potential adverse effects of ASMs on fetus development, and the effects of uncontrolled seizures on fetus and mother must be considered. In this scenario, seizure control prior to pregnancy represents the most important factor in predicting seizure control during pregnancy (Tomson et al., 2019). Valproate is associated with the highest risks of malformations, as well as adverse cognitive and behavioral outcomes, and should not be used as first line whenever possible in childnearing age women. The risk of major congenital malformations is dose-dependent for valproate and is probably also dose-dependent for other ASMs. Topiramate presents intermediate risk of malformation in specific organs. In contrast, lamotrigine and levetiracetam are associated with the lowest risks of malformations (Tomson et al., 2019). Prior to conception, it would be advisable a careful planning, both for the choice of an optimal ASM with little/no teratogenic potential, as well as for its dosage adjustment, and the initiation of folic acid supplementation prior to conception. During pregnancy, if the woman is taking an ASM that presents substantial changes in clearance (e.g. lamotrigine, levetiracetam and oxcarbazepine), monitoring of the drug level during pregnancy is recommended. Finally, several studies showed no adverse effects of breastfeeding when taking ASMs, therefore breastfeeding would be advisable (Tomson et al., 2019).

5.3 Neuropsychiatric comorbidities

Neuropsychiatric comorbidity is a particularly relevant aspect for both patients with epilepsy and brain tumors. Previously, a prevalence of neuropsychiatric disorders of 25–50% has been estimated among people with epilepsy (Lin et al., 2012), while a recent meta-analysis evidenced a prevalence of any mood disorder of 38.2% in oncology patients (Mitchell et al., 2011). Other studies have observed that up to two-thirds of patients with cancer and depression concomitantly present with anxiety symptoms (Smith, 2015). In neuro-oncological patients, especially with frontal-located tumors, prefrontal symptoms such as apathy, irritability, behavioral changes, or irascibility, should be closely evaluated. Moreover, neuropsychiatric comorbidity shows a relevant negative impact on the patient quality of life. It may be aggravated by some of the ASM used to treat BTRE-patients. Therefore, the ASM choice is highly impactful in this specific population.

5.4 Sudden unexpected death in epilepsy

Another important aspect to consider in BTRE-patients is sudden unexpected death in epilepsy (SUDEP). Nowadays, it is known that people with epilepsy have an increased risk of mortality compared to the general population, being higher in the first years of the disease, especially in those who are not treated with ASM (Hrabok et al., 2021; Kløvgaard et al., 2021). Other aspects that increase the risk of SUDEP are lack of adherence to treatment and poor seizure control, particularly when bilateral tonic-clonic seizures during sleep are present. Close monitoring and sleep video-EEG studies are mandatory to assess this relevant issue (DeGiorgio et al., 2019; Hrabok et al., 2021). Additionally, in patients at high risk of SUDEP, it is advisable to inform and empower both patient and family about the risk factors and ways to prevent it (Gutiérrez-Viedma et al., 2019).

5.5 Glioneural tumors

Glioneural neoplasms, such as disembryoplastic neuroepithelial tumors (DNETs) and gangliogliomas, constitute a specific group of tumors, as they represent highly epileptogenic developmental lesions characterized clinically by early onset of seizures and a tendency to drug resistance (Ertürk Çetin et al., 2017). The frequency of seizures reaches to almost 100% with DNETs and 80–90% with gangliogliomas (van Breemen et al., 2007). They are part of the group of “low-grade epilepsy-associated tumors” (LEATs). LEATs are a specific group of tumors strongly associated with epilepsy. Their characteristics include early-onset drug-resistant epilepsy, slow growth rate, neocortical localization, and temporal lobe predominance (Blümcke et al., 2016). Although DNET may be associated with focal cortical dysplasia, the impact of this on epileptogenicity is still unclear (Bonney et al., 2016). Generally, surgical resection is the corner stone of seizure management for patients with glioneuronal tumors (Krauze et al., 2015). Early surgical intervention and total macroscopic resection represent critically important factors in achieving seizure freedom and thus improving quality of life (Englot et al., 2012b).

6 Prognostic factors for seizure control in BTRE patients

There are several factors that may facilitate ASM resistance and prognosis in terms of seizure control. Thus, glioneural tumors (DNET and ganglioglioma) present highest rates of drug resistance. Among the main prognostic factors for seizure control after surgery are shorter duration of epilepsy (less than 1 year) and gross total resection (over subtotal lesionectomy) (Englot et al., 2012). In the case of low grade glial tumors, despite ASMs, approximately one-half of patients may be preoperatively drug-resistant with BTRE. Among some of the factors previously described, insular and/or parietal location of tumor lesions, history of epileptic seizure at diagnosis, and tumor within functional areas are factors associated with drug-resistant seizures (Pallud et al., 2014). Regarding treatment, the extent of resection was associated with improvement in post-treatment seizure control. (You et al., 2012; Pallud et al., 2014). Regarding high-grade gliomas, some works have highlighted that prolonged seizure control is associated with a better Karnofsky performance score, whereas uncontrolled preoperative seizures and parietal lobe involvement would be negative prognostic factors (Kerkhof et al., 2013).

7 Conclussion

The choice of the ASM in BTRE-patients is a complex decision determined by many factors. These include pharmacokinetic and pharmacodynamic characteristics, tolerability, efficacy, patient comorbidities, galenic formulations or clinician’s experience, among others. The choice of monotherapy versus polytherapy could be an optimal option to consider, given the minimization of pharmacological interactions. Subsequently, in case of failure to control epilepsy, a rational polytherapy with pharmacodynamic synergies could be an interesting option to consider. In general, ASMs with no (or less) hepatic enzyme induction or inhibition capacity such as levetiracetam, lacosamide, brivaracetam o perampanel would be preferable options to classical ASM given their greater drug-to-drug interactions. Some of the special situations to be considered would be patients with psychiatric comorbidity, elderly patients and women with reproductive desires or pregnancy. Finally, more studies will be needed to establish more optimal decisions on when, with what and until when to maintain ASMs in BTRE-patients.

Author contributions

JS-V: conceptualization, investigation, literature retrieval, writing—original draft and editing. AA-S: conceptualization, writing-review and supervision. IV-M and MFS: writing-review and supervision. MA: editing and supervision. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid; ASM, antiseizure medication; BM, brain metastasis; BTRE, brain metastasis-related epilepsy; DNET, disembryoplastic neuroepithelial tumors; GABA, gamma-aminobutyric acid; IDH, isocitrate dehydrogenase; LEATs, low-grade epilepsy-associated tumors; NMDA, N-methyl-d-aspartate; SCBs, sodium channel blockers; SRS, stereotactic radiosurgery; SV2a, synaptic vesicle protein 2a.

References

Alcántara-Montero, A., and Sánchez-Carnerero, C. I. (2016). Lacosamide and neuropathic pain, a review. Rev. Neurol. 62, 223–229. doi:10.33588/rn.6205.2015498

PubMed Abstract | CrossRef Full Text | Google Scholar

Aledo-Serrano, A., and Gil-Nagel, A. (2020). “Anticonvulsant agents: Carbamazepine, oxcarbazepine, and eslicarbazepine acetate,” in NeuroPsychopharmacotherapy. Editors R. Peter, L. Gerd, T. Nagatsu, W. Le, and R. Christian (Manhattan, New York: springerlink). doi:10.1007/978-3-319-56015-1

CrossRef Full Text | Google Scholar

Bähr, O., Hermisson, M., Rona, S., Rieger, J., Nussbaum, S., Körtvelyessy, P., et al. (2012). Intravenous and oral levetiracetam in patients with a suspected primary brain tumor and symptomatic seizures undergoing neurosurgery: The HELLO trial. Acta Neurochir. 154, 229–235. doi:10.1007/S00701-011-1144-9

CrossRef Full Text | Google Scholar

Baumgartner, T. R., and Elger, C. E. (2020). “Anti-convulsant agents: Valproic acid,” in NeuroPsychopharmacotherapy. Editors R. Peter, L. Gerd, T. Nagatsu, W. Le, and R. Christian (Manhattan, New York: springerlink), 1–9. doi:10.1007/978-3-319-56015-1_309-1

CrossRef Full Text | Google Scholar

Bedetti, C., Romoli, M., Maschio, M., Di Bonaventura, C., Nardi Cesarini, E., Eusebi, P., et al. (2017). Neuropsychiatric adverse events of antiepileptic drugs in brain tumour-related epilepsy: An Italian multicentre prospective observational study. Eur. J. Neurol. 24, 1283–1289. doi:10.1111/ene.13375

PubMed Abstract | CrossRef Full Text | Google Scholar

Beltrán-Corbellini, Á., Aledo-Serrano, Á., Møller, R. S., Pérez-Palma, E., García-Morales, I., Toledano, R., et al. (2022). Epilepsy genetics and precision medicine in adults: A new landscape for developmental and epileptic encephalopathies. Front. Neurol. 13, 777115. doi:10.3389/FNEUR.2022.777115

PubMed Abstract | CrossRef Full Text | Google Scholar

Berntsson, S. G., Merrell, R. T., Amirian, E. S., Armstrong, G. N., Lachance, D., Smits, A., et al. (2018). Glioma-related seizures in relation to histopathological subtypes: A report from the glioma international case-control study. J. Neurol. 265, 1432–1442. doi:10.1007/S00415-018-8857-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Bobustuc, G. C., Baker, C. H., Limaye, A., Jenkins, W. D., Pearl, G., Avgeropoulos, N. G., et al. (2010). Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro. Oncol. 12, 917–927. doi:10.1093/NEUONC/NOQ044

PubMed Abstract | CrossRef Full Text | Google Scholar

Bonney, P. A., Boettcher, L. B., Conner, A. K., Glenn, C. A., Briggs, R. G., Santucci, J. A., et al. (2016). Review of seizure outcomes after surgical resection of dysembryoplastic neuroepithelial tumors. J. Neurooncol. 126, 1–10. doi:10.1007/S11060-015-1961-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Bourg, V., Lebrun, C., Chichmanian, R. M., Thomas, P., and Frenay, M. (2001). Nitroso-urea–cisplatin-based chemotherapy associated with valproate: Increase of haematologic toxicity. Ann. Oncol. 12, 217–219. doi:10.1023/A:1008331708395

PubMed Abstract | CrossRef Full Text | Google Scholar

Bruin, De, Van Der Meer, P. B., Dirven, L., Taphoorn, M. J. B., and Koekkoek, J. A. F. (2021). Efficacy of antiepileptic drugs in glioma patients with epilepsy: A systematic review. Neurooncol. Pract. 8, 501–517. doi:10.1093/nop/npab030

PubMed Abstract | CrossRef Full Text | Google Scholar

Bruna, J., Miró, J., and Velasco, R. (2013). Epilepsy in glioblastoma patients: Basic mechanisms and current problems in treatment. Expert Rev. Clin. Pharmacol. 6, 333–344. doi:10.1586/ECP.13.12

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardona, A. F., Rojas, L., Wills, B., Bernal, L., Ruiz-Patiño, A., Arrieta, O., et al. (2018). Efficacy and safety of Levetiracetam vs. other antiepileptic drugs in Hispanic patients with glioblastoma. J. Neurooncol. 136, 363–371. doi:10.1007/S11060-017-2660-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Casas Parera, I., Roffo, G., Maria, A., Báez, A., Quintans, F., Castellanos Oropeza, P., et al. (2019). Characterization of seizures (ilae 1981 and 2017 classifications) and their response to treatment in a cohort of patients with glial tumors: A prospective single center study. eNeurologicalSci 14, 51–55. doi:10.1016/J.ENSCI.2018.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, D. Y., Chen, C. C., Crawford, J. R., and Wang, S. G. (2018). Tumor-related epilepsy: Epidemiology, pathogenesis and management. J. Neurooncol. 139, 13–21. doi:10.1007/s11060-018-2862-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Lusicic, A., O’Brien, T. J., Velakoulis, D., Adams, S. J., and Kwan, P. (2016). Psychotic disorders induced by antiepileptic drugs in people with epilepsy. Brain 139, 2668–2678. doi:10.1093/brain/aww196

PubMed Abstract | CrossRef Full Text | Google Scholar

Chonan, M., Saito, R., Kanamori, M., Osawa, S. I., Watanabe, M., Suzuki, H., et al. (2020). Experience of low dose perampanel to add-on in glioma patients with levetiracetam-uncontrollable epilepsy. Neurol. Med. Chir. 60, 37–44. doi:10.2176/NMC.OA.2018-0245

PubMed Abstract | CrossRef Full Text | Google Scholar

Contreras-García, I. J., Gómez-Lira, G., Phillips-Farfán, B. V., Pichardo-Macías, L. A., García-Cruz, M. E., Chávez-Pacheco, J. L., et al. (2021). Synaptic vesicle protein 2a expression in glutamatergic terminals is associated with the response to levetiracetam treatment. Brain Sci. 11, 531. doi:10.3390/brainsci11050531

PubMed Abstract | CrossRef Full Text | Google Scholar

Dannaram, S., Borra, D., Pulluri, M., Jindal, P., and Sharma, A. (2012). Levetiracetam-induced acute psychotic episode. Innov. Clin. Neurosci. 9, 10–12. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23198271 (Accessed November 21, 2020).

PubMed Abstract | Google Scholar

de Biase, S., Gigli, G. L., and Valente, M. (2020). Brivaracetam for the treatment of focal-onset seizures: Pharmacokinetic and pharmacodynamic evaluations. Expert Opin. Drug Metab. Toxicol. 16, 853–863. doi:10.1080/17425255.2020.1813277

PubMed Abstract | CrossRef Full Text | Google Scholar

De Groot, M., Aronica, E., Heimans, J. J., and Reijneveld, J. C. (2011). Synaptic vesicle protein 2A predicts response to levetiracetam in patients with glioma. Neurology 77, 532–539. doi:10.1212/WNL.0B013E318228C110

PubMed Abstract | CrossRef Full Text | Google Scholar

DeGiorgio, C. M., Curtis, A., Hertling, D., and Moseley, B. D. (2019). Sudden unexpected death in epilepsy: Risk factors, biomarkers, and prevention. Acta Neurol. Scand. 139, 220–230. doi:10.1111/ANE.13049

PubMed Abstract | CrossRef Full Text | Google Scholar

Dinapoli, L., Maschio, M., Jandolo, B., Fabi, A., Pace, A., Sperati, F., et al. (2009). Quality of life and seizure control in patients with brain tumor-related epilepsy treated with levetiracetam monotherapy: Preliminary data of an open-label study. Neurol. Sci. 30, 353–359. doi:10.1007/S10072-009-0087-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Dinkelacker, V., Dietl, T., Widman, G., Lengler, U., and Elger, C. E. (2003). Aggressive behavior of epilepsy patients in the course of levetiracetam add-on therapy: Report of 33 mild to severe cases. Epilepsy Behav. 4, 537–547. doi:10.1016/j.yebeh.2003.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Dreischmeier, E., Zuloaga, A., Kotloski, R. J., Karasov, A. O., and Gidal, B. E. (2021). Levetiracetam-associated irritability and potential role of vitamin B6 use in veterans with epilepsy. Epilepsy Behav. Rep. 16, 100452. doi:10.1016/J.EBR.2021.100452

PubMed Abstract | CrossRef Full Text | Google Scholar

Englot, D. J., Chang, E. F., and Vecht, C. J. (2016a). Epilepsy and brain tumors. Handb. Clin. Neurol. 134, 267–285. doi:10.1016/B978-0-12-802997-8.00016-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Englot, D. J., Magill, S. T., Han, S. J., Chang, E. F., Berger, M. S., and McDermott, M. W. (2016b). Seizures in supratentorial meningioma: A systematic review and meta-analysis. J. Neurosurg. 124, 1552–1561. doi:10.3171/2015.4.JNS142742

PubMed Abstract | CrossRef Full Text | Google Scholar

Ertürk Çetin, Ö., İşler, C., Uzan, M., and Özkara, Ç. (2017). Epilepsy-related brain tumors. Seizure 44, 93–97. doi:10.1016/j.seizure.2016.12.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Feyissa, A. M. (2019). Brivaracetam in the treatment of epilepsy: A review of clinical trial data. Neuropsychiatr. Dis. Treat. 15, 2587–2600. doi:10.2147/NDT.S143548

PubMed Abstract | CrossRef Full Text | Google Scholar

Fox, J., Ajinkya, S., Houston, P., Lindhorst, S., Cachia, D., Olar, A., et al. (2019). Seizures in patents with primary central nervous system lymphoma: Prevalence and associated features. J. Neurol. Sci. 400, 34–38. doi:10.1016/J.JNS.2019.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Glantz, M. J., Cole, B. F., Forsyth, P. A., Recht, L. D., Wen, P. Y., Chamberlain, M. C., et al. (2000). Practice parameter: Anticonvulsant prophylaxis in patients with newly diagnosed brain tumors: Report of the quality standards subcommittee of the American academy of Neurology. Neurology 54, 1886–1893. doi:10.1212/WNL.54.10.1886

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldstein, E. D., and Feyissa, A. M. (2018). Brain tumor related-epilepsy. Neurol. Neurochir. Pol. 52, 436–447. doi:10.1016/j.pjnns.2018.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Greenhalgh, J., Weston, J., Dundar, Y., Nevitt, S. J., and Marson, A. G. (2020). Antiepileptic drugs as prophylaxis for postcraniotomy seizures. Cochrane Database Syst. Rev. 4, CD007286. doi:10.1002/14651858.CD007286.PUB52020

PubMed Abstract | CrossRef Full Text | Google Scholar

Grupo Español de Investigación en Neurooncología (2021). Manejo y tratamiento de crisis epilépticas en pacientes con tumores cerebrales y cánceres sistémicos, 1–9.

Google Scholar

Gutiérrez-Viedma, Á., Sanz-Graciani, I., Romeral-Jiménez, M., Parejo-Carbonell, B., Serrano-García, I., Cuadrado, M. L., et al. (2019). Patients’ knowledge on epilepsy and SUDEP improves after a semi-structured health interview. Epilepsy Behav. 99, 106467. doi:10.1016/J.YEBEH.2019.106467

PubMed Abstract | CrossRef Full Text | Google Scholar

Haggiagi, A., and Avila, E. K. (2019). Seizure response to temozolomide chemotherapy in patients with WHO grade II oligodendroglioma: A single-institution descriptive study. Neurooncol. Pract. 6, 203–208. doi:10.1093/NOP/NPY029

PubMed Abstract | CrossRef Full Text | Google Scholar

Happold, C., Gorlia, T., Chinot, O., Gilbert, M. R., Nabors, L. B., Wick, W., et al. (2016). 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. doi:10.1200/JCO.2015.63.6563

PubMed Abstract | CrossRef Full Text | Google Scholar

Hovinga, C. A. (2001). Levetiracetam: A novel antiepileptic drug. Pharmacotherapy 21, 1375–1388. doi:10.1592/phco.21.17.1375.34432

PubMed Abstract | CrossRef Full Text | Google Scholar

Hrabok, M., Engbers, J. D. T., Wiebe, S., Sajobi, T. T., Subota, A., Almohawes, A., et al. (2021). Primary care electronic medical records can be used to predict risk and identify potentially modifiable factors for early and late death in adult onset epilepsy. Epilepsia 62, 51–60. doi:10.1111/EPI.16738

PubMed Abstract | CrossRef Full Text | Google Scholar

Italiano, D., and Perucca, E. (2013). Clinical pharmacokinetics of new-generation antiepileptic drugs at the extremes of age: An update. Clin. Pharmacokinet. 52, 627–645. doi:10.1007/S40262-013-0067-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Iuchi, T., Hasegawa, Y., Kawasaki, K., and Sakaida, T. (2015). Epilepsy in patients with gliomas: Incidence and control of seizures. J. Clin. Neurosci. 22, 87–91. doi:10.1016/j.jocn.2014.05.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Ius, T., Pauletto, G., Tomasino, B., Maieron, M., Budai, R., Isola, M., et al. (2020). Predictors of postoperative seizure outcome in low grade glioma: From volumetric analysis to molecular stratification. Cancers (Basel) 12, E397. doi:10.3390/CANCERS12020397

PubMed Abstract | CrossRef Full Text | Google Scholar

Izumoto, S., Miyauchi, M., Tasaki, T., Okuda, T., Nakagawa, N., Nakano, N., et al. (2018). Seizures and tumor progression in glioma patients with uncontrollable epilepsy treated with perampanel. Anticancer Res. 38, 4361–4366. doi:10.21873/ANTICANRES.12737

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanner, A. M. (2016a). Management of psychiatric and neurological comorbidities in epilepsy. Nat. Rev. Neurol. 12, 106–116. doi:10.1038/nrneurol.2015.243

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanner, A. M., and Bicchi, M. M. (2022). Antiseizure medications for adults with epilepsy: A review. JAMA 327, 1269–1281. doi:10.1001/JAMA.2022.3880

PubMed Abstract | CrossRef Full Text | Google Scholar

Kellinghaus, C. (2009). Lacosamide as treatment for partial epilepsy: Mechanisms of action, pharmacology, effects, and safety. Ther. Clin. Risk Manag. 5, 757. doi:10.2147/tcrm.s5189

PubMed Abstract | CrossRef Full Text | Google Scholar

Kerkhof, M., Dielemans, J. C. M., Breemen, Van, Melanie, S., Zwinkels, H., Walchenbach, R., et al. (2013). Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro. Oncol. 15, 961–967. doi:10.1093/NEUONC/NOT057

PubMed Abstract | CrossRef Full Text | Google Scholar

Kerkhof, M., Koekkoek, J. A. F., Vos, M. J., van den Bent, M. J., Taal, W., Postma, T. J., et al. (2019). Withdrawal of antiepileptic drugs in patients with low grade and anaplastic glioma after long-term seizure freedom: A prospective observational study. J. Neurooncol. 142, 463–470. doi:10.1007/S11060-019-03117-Y

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y. H., Kim, T., Joo, J. D., Han, J. H., Kim, Y. J., Kim, I. A., et al. (2015). Survival benefit of levetiracetam in patients treated with concomitant chemoradiotherapy and adjuvant chemotherapy with temozolomide for glioblastoma multiforme. Cancer 121, 2926–2932. doi:10.1002/CNCR.29439

PubMed Abstract | CrossRef Full Text | Google Scholar

Klitgaard, H., Matagne, A., Gobert, J., and Wülfert, E. (1998). Evidence for a unique profile of levetiracetam in rodent models of seizures and epilepsy. Eur. J. Pharmacol. 353, 191–206. doi:10.1016/S0014-2999(98)00410-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Kløvgaard, M., Lynge, T. H., Tsiropoulos, I., Uldall, P. V., Banner, J., Winkel, B. G., et al. (2021). Sudden unexpected death in epilepsy in persons younger than 50 years: A retrospective nationwide cohort study in Denmark. Epilepsia 62, 2405–2415. doi:10.1111/EPI.17037

PubMed Abstract | CrossRef Full Text | Google Scholar

Krauze, A. V., Myrehaug, S. D., Chang, M. G., Holdford, D. J., Smith, S., Shih, J., et al. (2015). A phase 2 study of concurrent radiation therapy, temozolomide, and the histone deacetylase inhibitor valproic acid for patients with glioblastoma. Int. J. Radiat. Oncol. Biol. Phys. 92, 986–992. doi:10.1016/J.IJROBP.2015.04.038

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuijlen, J. M., Teernstra, O. P., Kessels, A. G., Herpers, M. J., and Beuls, E. A. (1996). Effectiveness of antiepileptic prophylaxis used with supratentorial craniotomies: A meta-analysis. Seizure 5, 291–298. doi:10.1016/s1059-1311(96)80023-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, J. W., Wen, P. Y., Hurwitz, S., Black, P., Kesari, S., Drappatz, J., et al. (2010). Morphological characteristics of brain tumors causing seizures. Arch. Neurol. 67, 336–342. doi:10.1001/archneurol.2010.2

PubMed Abstract | CrossRef Full Text | Google Scholar

Leslie, T. K., Brückner, L., Chawla, S., and Brackenbury, W. J. (2020). Inhibitory effect of eslicarbazepine acetate and S-licarbazepine on Nav1.5 channels. Front. Pharmacol. 11, 555047. doi:10.3389/fphar.2020.555047.2020.555047/BIBTEX

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, J. J., Mula, M., and Hermann, B. P. (2012). Uncovering the neurobehavioural comorbidities of epilepsy over the lifespan. Lancet 380, 1180–1192. doi:10.1016/S0140-6736(12)61455-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Löscher, W., and Klein, P. (2021). The pharmacology and clinical efficacy of antiseizure medications: From bromide salts to cenobamate and beyond. CNS Drugs 35, 935–963. doi:10.1007/S40263-021-00827-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, Y., Yu, W., and Wang, X. (2009). Efficacy of topiramate in adult patients with symptomatic epilepsy: An open-label, long-term, retrospective observation. CNS Drugs 23, 351–359. doi:10.2165/00023210-200923040-00006

PubMed Abstract | CrossRef Full Text | Google Scholar

Lynch, B. A., Lambeng, N., Nocka, K., Kensel-Hammes, P., Bajjalieh, S. M., Matagne, A., et al. (2004). The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc. Natl. Acad. Sci. U. S. A. 101, 9861–9866. doi:10.1073/pnas.0308208101

PubMed Abstract | CrossRef Full Text | Google Scholar

Marino, S., Vitaliti, G., Marino, S. D., Pavone, P., Provvidenti, S., Romano, C., et al. (2018). Pyridoxine add-on treatment for the control of behavioral adverse effects induced by levetiracetam in children: A case-control prospective study. Ann. Pharmacother. 52, 645–649. doi:10.1177/1060028018759637

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M. (2012). Brain tumor-related epilepsy. Curr. Neuropharmacol. 10, 124–133. doi:10.2174/157015912800604470

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Albani, F., Baruzzi, A., Zarabla, A., Dinapoli, L., Pace, A., et al. (2006). Levetiracetam therapy in patients with brain tumour and epilepsy. J. Neurooncol. 80, 97–100. doi:10.1007/s11060-006-9162-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Mingoia, M., Sperati, F., Pace, A., Pompili, A., et al. (2011a). Lacosamide as add-on in brain tumor-related epilepsy: Preliminary report on efficacy and tolerability. J. Neurol. 258, 2100–2104. doi:10.1007/s00415-011-6132-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Sperati, F., Fabi, A., Pace, A., Vidiri, A., et al. (2012a). 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. doi:10.1007/S11060-011-0689-Z

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Sperati, F., Pace, A., Fabi, A., Vidiri, A., et al. (2012b). Effect of pregabalin add-on treatment on seizure control, quality of life, and anxiety in patients with brain tumour-related epilepsy: A pilot study. Epileptic Disord. 14, 388–397. doi:10.1684/EPD.2012.0542

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Sperati, F., Pace, A., Fabi, A., Vidiri, A., et al. (2011b). Levetiracetam monotherapy in patients with brain tumor-related epilepsy: Seizure control, safety, and quality of life. J. Neurooncol. 104, 205–214. doi:10.1007/S11060-010-0460-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Vidiri, A., Pace, A., Fabi, A., Pompili, A., et al. (2009). The role side effects play in the choice of antiepileptic therapy in brain tumor-related epilepsy: A comparative study on traditional antiepileptic drugs versus oxcarbazepine. J. Exp. Clin. Cancer Res. 28, 60. doi:10.1186/1756-9966-28-60

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Zarabla, A., Maialetti, A., Giannarelli, D., Fabi, A., et al. (2017a). Zonisamide in brain tumor-related epilepsy: An observational pilot study. Clin. Neuropharmacol. 40, 113–119. doi:10.1097/WNF.0000000000000218

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Dinapoli, L., Zarabla, A., Pompili, A., Carapella, C. M., Pace, A., et al. (2008). Outcome and tolerability of topiramate in brain tumor associated epilepsy. J. Neurooncol. 86, 61–70. doi:10.1007/S11060-007-9430-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Maialetti, A., Mocellini, C., Domina, E., Pauletto, G., Costa, C., et al. (2020a). Effect of brivaracetam on efficacy and tolerability in patients with brain tumor-related epilepsy: A retrospective multicenter study. Front. Neurol. 11, 813–817. doi:10.3389/fneur.2020.00813

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Pauletto, G., Zarabla, A., Maialetti, A., Lus, T., Villani, V., et al. (2019). Perampanel in patients with brain tumor-related epilepsy in real-life clinical practice: A retrospective analysis. Int. J. Neurosci. 129, 593–597. doi:10.1080/00207454.2018.1555160

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Zarabla, A., Maialetti, A., Fabi, A., Vidiri, A., Villani, V., et al. (2017b). Quality of life, mood and seizure control in patients with brain tumor related epilepsy treated with lacosamide as add-on therapy: A prospective explorative study with a historical control group. Epilepsy Behav. 73, 83–89. doi:10.1016/j.yebeh.2017.05.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Maschio, M., Zarabla, A., Maialetti, A., Giannarelli, D., Koudriavtseva, T., Villani, V., et al. (2020b). Perampanel in brain tumor-related epilepsy: Observational pilot study. Brain Behav. 10, e01612. doi:10.1002/BRB3.1612

PubMed Abstract | CrossRef Full Text | Google Scholar

Merrell, R. T., Anderson, S. K., Meyer, F. B., and Lachance, D. H. (2010). Seizures in patients with glioma treated with phenytoin and levetiracetam. J. Neurosurg. 113, 1176–1181. doi:10.3171/2010.5.JNS091367

PubMed Abstract | CrossRef Full Text | Google Scholar

Michelucci, R., Pasini, E., Meletti, S., Fallica, E., Rizzi, R., Florindo, I., et al. (2013). Epilepsy in primary cerebral tumors: The characteristics of epilepsy at the onset (results from the PERNO study – project of Emilia Romagna Region on Neuro-Oncology). Epilepsia 54, 86–91. doi:10.1111/EPI.12314

PubMed Abstract | CrossRef Full Text | Google Scholar

Minniti, G., Lombardi, G., and Paolini, S. (2019). Glioblastoma in elderly patients: Current management and future perspectives. Cancers 11, E336. doi:10.3390/CANCERS11030336

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitchell, A. J., Chan, M., Bhatti, H., Halton, M., Grassi, L., Johansen, C., et al. (2011). Prevalence of depression, anxiety, and adjustment disorder in oncological, haematological, and palliative-care settings: A meta-analysis of 94 interview-based studies. Lancet. Oncol. 12, 160–174. doi:10.1016/S1470-2045(11)70002-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Mo, F., Meletti, S., Belcastro, V., Quadri, S., Napolitano, M., Bello, L., et al. (2022). Lacosamide in monotherapy in BTRE (brain tumor-related epilepsy): Results from an Italian multicenter retrospective study. J. Neurooncol. 157, 551–559. doi:10.1007/S11060-022-03998-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Newton, H. B., Goldlust, S. A., and Pearl, D. (2006). Retrospective analysis of the efficacy and tolerability of levetiracetam in brain tumor patients. J. Neurooncol. 78, 99–102. doi:10.1007/s11060-005-9070-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Niespodziany, I., Rigo, J. M., Moonen, G., Matagne, A., Klitgaard, H., and Wolff, C. (2017). Brivaracetam does not modulate ionotropic channels activated by glutamate, γ-aminobutyric acid, and glycine in hippocampal neurons. Epilepsia 58, e157–e161. doi:10.1111/epi.13890

PubMed Abstract | CrossRef Full Text | Google Scholar

Pallud, J., Audureau, E., Blonski, M., Sanai, N., Bauchet, L., Fontaine, D., et al. (2014). Epileptic seizures in diffuse low-grade gliomas in adults. Brain 137, 449–462. doi:10.1093/BRAIN/AWT345

PubMed Abstract | CrossRef Full Text | Google Scholar

Pallud, J., Huberfeld, G., Dezamis, E., Peeters, S., Moiraghi, A., Gavaret, M., et al. (2021). Effect of levetiracetam use duration on overall survival of isocitrate dehydrogenase wild-type glioblastoma in adults. Neurology 98, e125–e140. doi:10.1212/WNL.0000000000013005

PubMed Abstract | CrossRef Full Text | Google Scholar

Perucca, E., and Tomson, T. (2011). The pharmacological treatment of epilepsy in adults. Lancet. Neurol. 10, 446–456. doi:10.1016/S1474-4422(11)70047-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Pourzitaki, C., Tsaousi, G., Apostolidou, E., Karakoulas, K., Kouvelas, D., and Amaniti, E. (2016). Efficacy and safety of prophylactic levetiracetam in supratentorial brain tumour surgery: A systematic review and meta-analysis. Br. J. Clin. Pharmacol. 82, 315–325. doi:10.1111/bcp.12926

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizzo, A., Donzelli, S., Girgenti, V., Sacconi, A., Vasco, C., Salmaggi, A., et al. (2017). In vitro antineoplastic effects of brivaracetam and lacosamide on human glioma cells. J. Exp. Clin. Cancer Res. 36, 76. doi:10.1186/S13046-017-0546-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Rocamora, R., Álvarez, I., Chavarría, B., and Principe, A. (2020). Perampanel effect on sleep architecture in patients with epilepsy. Seizure 76, 137–142. doi:10.1016/J.SEIZURE.2020.01.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Rosati, A., Buttolo, L., Stefini, R., Todeschini, A., Cenzato, M., and Padovani, A. (2010). Efficacy and safety of levetiracetam in patients with glioma: A clinical prospective study. Arch. Neurol. 67, 343–346. doi:10.1001/ARCHNEUROL.2009.335

PubMed Abstract | CrossRef Full Text | Google Scholar

Rossetti, A. O., Jeckelmann, S., Novy, J., Roth, P., Weller, M., and Stupp, R. (2014). Levetiracetam and pregabalin for antiepileptic monotherapy in patients with primary brain tumors. A phase II randomized study. Neuro. Oncol. 16, 584–588. doi:10.1093/NEUONC/NOT170

PubMed Abstract | CrossRef Full Text | Google Scholar

Rudà, R., Bello, L., Duffau, H., and Soffietti, R. (2012). Seizures in low-grade gliomas: Natural history, pathogenesis, and outcome after treatments. Neuro. Oncol. 14 (14), iv55–iv64. doi:10.1093/NEUONC/NOS199

PubMed Abstract | CrossRef Full Text | Google Scholar

Rudà, R., Houillier, C., Maschio, M., Reijneveld, J. C., Hellot, S., De Backer, M., et al. (2020). Effectiveness and tolerability of lacosamide as add-on therapy in patients with brain tumor-related epilepsy: Results from a prospective, noninterventional study in European clinical practice (VIBES). Epilepsia 61, 647–656. doi:10.1111/epi.16486

PubMed Abstract | CrossRef Full Text | Google Scholar

Rudà, R., Pellerino, A., Franchino, F., Bertolotti, C., Bruno, F., Mo, F., et al. (2018). Lacosamide in patients with gliomas and uncontrolled seizures: Results from an observational study. J. Neurooncol. 136, 105–114. doi:10.1007/s11060-017-2628-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Salmaggi, A., Corno, C., Maschio, M., Donzelli, S., D’urso, A., Perego, P., et al. (2021). Synergistic effect of perampanel and temozolomide in human glioma cell lines. J. Pers. Med. 390 (11), 390. doi:10.3390/JPM11050390

CrossRef Full Text | Google Scholar

Sánchez-Villalobos, J. M., Aledo-Serrano, Á., Serna-Berna, A., Salinas-Ramos, J., Martínez-Alonso, E., Pérez-Vicente, J. A., et al. (2021a). Antiseizure medication for brain metastasis-related epilepsy: Findings of optimal choice from a retrospective cohort. Epilepsy Res. 178, 106812. doi:10.1016/j.eplepsyres.2021.106812

CrossRef Full Text | Google Scholar

Sánchez-Villalobos, J. M., Serna-Berna, A., Salinas-Ramos, J., Escolar-Pérez, P. P., Martínez-Alonso, E., Achel, D. G., et al. (2021b). Volumetric modulated arc radiosurgery for brain metastases from breast cancer: A single-center study. Colomb. Med. 52, e2004567. doi:10.25100/cm.v52i3.4567

CrossRef Full Text | Google Scholar

Sánchez-Villalobos, J. M., Villegas-Martínez, I., and Pérez-Vicente, J. A. (2018). A well-tolerated and effective antiepileptic drug for patients with myasthenia gravis at last? Clin. Neuropharmacol. 41, 80–81. doi:10.1097/WNF.0000000000000267

PubMed Abstract | CrossRef Full Text | Google Scholar

Santamarina, E., Parejo Carbonell, B., Sala, J., Gutiérrez-Viedma, Á., Miró, J., Asensio, M., et al. (2019). Use of intravenous brivaracetam in status epilepticus: A multicenter registry. Epilepsia 60, 1593–1601. doi:10.1111/EPI.16094

PubMed Abstract | CrossRef Full Text | Google Scholar

Saria, M. G., Corle, C., Hu, J., Rudnick, J. D., Phuphanich, S., Mrugala, M. M., et al. (2013). Retrospective analysis of the tolerability and activity of lacosamide in patients with brain tumors: Clinical article. J. Neurosurg. 118, 1183–1187. doi:10.3171/2013.1.JNS12397

PubMed Abstract | CrossRef Full Text | Google Scholar

Seo, J. G., Cho, Y. W., Kim, K. T., Kim, D. W., Yang, K. I., Lee, S. T., et al. (2020). Pharmacological treatment of epilepsy in elderly patients. J. Clin. Neurol. 16, 556–561. doi:10.3988/JCN.2020.16.4.556

PubMed Abstract | CrossRef Full Text | Google Scholar

Simó, M., Velasco, R., Graus, F., Verger, E., Gil, M., Pineda, E., et al. (2012). Impact of antiepileptic drugs on thrombocytopenia in glioblastoma patients treated with standard chemoradiotherapy. J. Neurooncol. 108, 451–458. doi:10.1007/S11060-012-0836-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, R., Stoltzfus, K. C., Chen, H., Louie, A. V., Lehrer, E. J., Horn, S. R., et al. (2020). Epidemiology of synchronous brain metastases. Neurooncol. Adv. 2, vdaa041–10. doi:10.1093/NOAJNL/VDAA041

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith, H. R. (2015). Depression in cancer patients: Pathogenesis, implications and treatment (Review). Oncol. Lett. 9, 1509–1514. doi:10.3892/ol.2015.2944

PubMed Abstract | CrossRef Full Text | Google Scholar

Solomons, M. R., Jaunmuktane, Z., Weil, R. S., El-Hassan, T., Brandner, S., and Rees, J. H. (2020). Seizure outcomes and survival in adult low-grade glioma over 11 years: Living longer and better. Neurooncol. Pract. 7, 196–201. doi:10.1093/NOP/NPZ056

PubMed Abstract | CrossRef Full Text | Google Scholar

Stöhr, T., Krause, E., and Selve, N. (2006). Lacosamide displays potent antinociceptive effects in animal models for inflammatory pain. Eur. J. Pain 10, 241–249. doi:10.1016/j.ejpain.2005.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Strzelczyk, A., Zöllner, J. P., Willems, L. M., Jost, J., Paule, E., Schubert-Bast, S., et al. (2017). Lacosamide in status epilepticus: Systematic review of current evidence. Epilepsia 58, 933–950. doi:10.1111/epi.13716

PubMed Abstract | CrossRef Full Text | Google Scholar

Thelengana, A., Shukla, G., Srivastava, A., Singh, M. B., Gupta, A., Rajan, R., et al. (2019). Cognitive, behavioural and sleep-related adverse effects on introduction of levetiracetam versus oxcarbazepine for epilepsy. Epilepsy Res. 150, 58–65. doi:10.1016/j.eplepsyres.2019.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Tomson, T., Battino, D., Bromley, R., Kochen, S., Meador, K., Pennell, P., et al. (2019). Executive summary: Management of epilepsy in pregnancy: A report from the international league against epilepsy task force on women and pregnancy. Epilepsia 00, 2343–2345. doi:10.1111/epi.16395

PubMed Abstract | CrossRef Full Text | Google Scholar

Usery, J. B., Michael, L. M., Sills, A. K., and Finch, C. K. (2010). A prospective evaluation and literature review of levetiracetam use in patients with brain tumors and seizures. J. Neurooncol. 99, 251–260. doi:10.1007/S11060-010-0126-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Breemen, M. S. M., Rijsman, R. M., Taphoorn, M. J. B., Walchenbach, R., Zwinkels, H., and Vecht, C. J. (2009). Efficacy of anti-epileptic drugs in patients with gliomas and seizures. J. Neurol. 256, 1519–1526. doi:10.1007/S00415-009-5156-9

PubMed Abstract | CrossRef Full Text | Google Scholar

van Breemen, M. S. M., Wilms, E. B., and Vecht, C. J. (2007). Epilepsy in patients with brain tumours: Epidemiology, mechanisms, and management. Lancet. Neurol. 6, 421–430. doi:10.1016/S1474-4422(07)70103-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Vecht, C., Duran-Peña, A., Houillier, C., Durand, T., Capelle, L., and Huberfeld, G. (2017). Seizure response to perampanel in drug-resistant epilepsy with gliomas: Early observations. J. Neurooncol. 133, 603–607. doi:10.1007/S11060-017-2473-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Villanueva, V., López-González, F. J., Mauri, J. A., Rodriguez-Uranga, J., Olivé-Gadea, M., Montoya, J., et al. (2019). BRIVA-LIFE–A multicenter retrospective study of the long-term use of brivaracetam in clinical practice. Acta Neurol. Scand. 139, 360–368. doi:10.1111/ane.13059

PubMed Abstract | CrossRef Full Text | Google Scholar

Villanueva, V., Saiz-Diaz, R., Toledo, M., Piera, A., Mauri, J. A., Rodriguez-Uranga, J. J., et al. (2016). NEOPLASM study: Real-life use of lacosamide in patients with brain tumor-related epilepsy. Epilepsy Behav. 65, 25–32. doi:10.1016/j.yebeh.2016.09.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Wagner, G., Eb, W., Van, D., and ChJ, V. (2003). Levetiracetam: Preliminary experience in patients with primary brain tumours. Seizure 12, 585. doi:10.1016/S1059-1311(03)00096-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Warnke, P. C., Berlis, A., Weyerbrock, A., and Ostertag, C. B. (1997). Significant reduction of seizure incidence and increase of benzodiazepine receptor density after interstitial radiosurgery in low-grade gliomas. Acta Neurochir. Suppl. 68, 90–92. doi:10.1007/978-3-7091-6513-3_17

PubMed Abstract | CrossRef Full Text | Google Scholar

Weintraub, D., Buchsbaum, R., Resor, S. R., and Hirsch, L. J. (2007). Psychiatric and behavioral side effects of the newer antiepileptic drugs in adults with epilepsy. Epilepsy Behav. 10, 105–110. doi:10.1016/J.YEBEH.2006.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Weller, M., Gorlia, T., Cairncross, J. G., Van Den Bent, M. J., Mason, W., Belanger, K., et al. (2011). Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma. Neurology 77, 1156–1164. doi:10.1212/WNL.0B013E31822F02E1

PubMed Abstract | CrossRef Full Text | Google Scholar

Wick, W., Menn, O., Meisner, C., Steinbach, J., Hermisson, M., Tatagiba, M., et al. (2005). Pharmacotherapy of epileptic seizures in glioma patients: Who, when, why and how long? Onkologie 28, 391–396. doi:10.1159/000086375

PubMed Abstract | CrossRef Full Text | Google Scholar

Wirsching, H. G., Morel, C., Gmür, C., Neidert, M. C., Baumann, C. R., Valavanis, A., et al. (2016). Predicting outcome of epilepsy after meningioma resection. Neuro. Oncol. 18, 1002–1010. doi:10.1093/NEUONC/NOV303

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolpert, F., Lareida, A., Terziev, R., Grossenbacher, B., Neidert, M. C., Roth, P., et al. (2020). Risk factors for the development of epilepsy in patients with brain metastases. Neuro. Oncol. 22, 718–728. doi:10.1093/NEUONC/NOZ172

PubMed Abstract | CrossRef Full Text | Google Scholar

Wright, C., Downing, J., Mungall, D., Khan, O., Williams, A., Fonkem, E., et al. (2013). Clinical pharmacology and pharmacokinetics of levetiracetam. Front. Neurol. 4, 192–196. doi:10.3389/fneur.2013.00192

PubMed Abstract | CrossRef Full Text | Google Scholar

You, G., Sha, Z. Y., Yan, W., Zhang, W., Wang, Y. Z., Li, S. W., et al. (2012). Seizure characteristics and outcomes in 508 Chinese adult patients undergoing primary resection of low-grade gliomas: A clinicopathological study. Neuro. Oncol. 14, 230–241. doi:10.1093/NEUONC/NOR205

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, Y., Xiang, W., Yanhui, L., Ruofei, L., Shuang, L., Yingjun, F., et al. (2013). Ki-67 overexpression in WHO grade II gliomas is associated with poor postoperative seizure control. Seizure 22, 877–881. doi:10.1016/J.SEIZURE.2013.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Zaatreh, M. M., Firlik, K. S., Spencer, D. D., and Spencer, S. S. (2003). Temporal lobe tumoral epilepsy: Characteristics and predictors of surgical outcome. Neurology 61, 636–641. doi:10.1212/01.WNL.0000079374.78589.1B

PubMed Abstract | CrossRef Full Text | Google Scholar

Zaatreh, M. M., Spencer, D. D., Thompson, J. L., Blumenfeld, H., Novotny, E. J., Mattson, R. H., et al. (2002). Frontal lobe tumoral epilepsy: Clinical, neurophysiologic features and predictors of surgical outcome. Epilepsia 43, 727–733. doi:10.1046/J.1528-1157.2002.39501.X

PubMed Abstract | CrossRef Full Text | Google Scholar

Zoccarato, M., Basile, A. M., Padovan, M., Caccese, M., Zagonel, V., and Lombardi, G. (2021). Eslicarbazepine in patients with brain tumor-related epilepsy: A single-center experience. Int. J. Neurosci. 131, 879–884. doi:10.1080/00207454.2020.1759590

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: antiseizure medication, brain tumor, glial tumor, seizure, sodium channel blockers, precision medicine

Citation: Sánchez-Villalobos JM, Aledo-Serrano Á, Villegas-Martínez I, Shaikh MF and Alcaraz M (2022) Epilepsy treatment in neuro-oncology: A rationale for drug choice in common clinical scenarios. Front. Pharmacol. 13:991244. doi: 10.3389/fphar.2022.991244

Received: 11 July 2022; Accepted: 14 September 2022;
Published: 06 October 2022.

Edited by:

Rita Citraro, University Magna Graecia of Catanzaro, Italy

Reviewed by:

Jyotirmoy Banerjee, All India Institute of Medical Sciences, India

Copyright © 2022 Sánchez-Villalobos, Aledo-Serrano, Villegas-Martínez, Shaikh and Alcaraz. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ángel Aledo-Serrano, aaledo@neurologiaclinica.es

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.