Abstract
There is increasing recognition that epilepsy can be associated with a broad spectrum of comorbidities. While epileptic seizures are an essential element of epilepsy in children, there is a spectrum of neurological, mental health and cognitive disorders that add to the disease burden of childhood epilepsy resulting in a decreased quality of life.
The most common comorbid conditions in childhood epilepsy include depression, anxiety, autism spectrum disorders, sleep disorders, attention deficits, cognitive impairment and migraine. While epilepsy can result in comorbidities, many of the comorbidities of childhood have a bi-directional association, with the comorbid condition increasing risk for epilepsy and epilepsy increasing the risk for the comorbid condition. The bidirectional feature of epilepsy and the comorbidities suggest a common underlying pathological basis for both the seizures and comorbid condition.
While recognition of the comorbid conditions of pediatric epilepsies is increasing, there has been a lag in the development of effective therapies partly out of concern that drugs used to treat the comorbid conditions could increase seizure susceptibility. There is now some evidence that most drugs used for comorbid conditions are safe and do not lower seizure threshold. Unfortunately, the evidence showing drugs are effective in treating many of the childhood comorbidities of epilepsy is quite limited. There is a great need for randomized, placebo-controlled drug trials for efficacy and safety in the treatment of comorbidities of childhood epilepsy.
1. Introduction
While epileptic seizures are the quintessential defining element of epilepsy, there has been increasing recognition that there is a spectrum of cognitive, behavioral, and psychiatric disorders that accompany epilepsy [1, 2]. Reflecting the idea that epilepsy can be associated with a broad spectrum of comorbidities, epilepsy is now defined as “a disease characterized by an enduring predisposition to generate epileptic seizures and by the neurobiological, cognitive, psychological, and social consequences of this condition” [3]. Indeed, comorbid conditions, defined as a greater than coincidental association of two conditions in the same individual [4], may be more problematic than the seizures, greatly impacting quality of life [5-8]. The additional cost and burden on the health care system of comorbidities associated with epilepsy, while difficult to quantify, are likely to be substantial [9].
Prominent comorbidities among children with prevalent epilepsy include neurological disorders, mental health conditions and cognitive disorders (Table 1) [9]. The relationship between comorbid conditions and epilepsy is complex (Figure 1 and Figure 2). In some conditions, epilepsy can result in the comorbid condition. For example, epilepsy can lead to an anxiety disorder or sleep disorder. In other situations, disorders such as brain neoplasms can result in epilepsy. Effectively treating the epilepsy in the first condition could reduce or eliminate the comorbid conditions whereas, in the second scenario, treating the cause of the epilepsy could eliminate the epilepsy. Rather than a cause-effect, in other comorbid conditions a common pathophysiological substrate leading to both conditions is likely [10-12]. For instance, attention deficit hyperactive disorder (ADHD), autism spectrum disorder (ASD), depression and migraine are risk factors for epilepsy and epilepsy is a risk factor for ADHD, ASD, depression, sleep disorders and migraine. In these cases, effective treatment of the epilepsy would not necessarily treat the comorbid condition. However, targeting the pathophysiological mechanism common to both conditions with one drug could conceivably treat both the epilepsy and comorbid condition.
Table 1.
Common comorbid conditions in children with epilepsy [9].
| Neurological disorders | Brain neoplasm Autism spectrum disorder Cerebral palsy Migraine Sleep disorders |
|---|---|
| Mental health conditions | Mood disorders (depression) Anxiety disorders Attention deficit hyperactivity disorders (ADHD) |
| Cognitive disorders | Intellectual disability Learning disability Memory impairment Executive dysfunction |
Figure 1.
Comorbidity paradigm. The relationship between epilepsy and comorbidities. Brain disorders can result in both epilepsy and comorbidities including cognitive impairment, ASD depression/anxiety, sleep disturbances, ADHD and migraines. Ongoing epileptic seizures can exacerbate comorbidities and epilepsy comorbidities can increase risk of seizures. Many comorbidities are bidirectional with epilepsy with the comorbidities occurring before the onset of the seizures. The bidirectionality of epilepsy and comorbidities suggest a common underlying pathogenetic mechanism in some children. In these cases, targeting the underlying mechanism of both conditions has the potential of reducing both the epilepsy and comorbidity.
Figure 2.
Schematic of relationship between epilepsy and AED therapy and common comorbidities of epilepsy in children. Children with epilepsy have a high risk for cognitive impairment, ASD depression/anxiety, sleep disturbances, ADHD and migraines. There is bidirectionality between epilepsy and the comorbidities. In addition, there is also a relationship between all of the epilepsy comorbidities as designated by the external circle.
Assessment and treatment of the comorbidities can be challenging. The clinical presentations of comorbid conditions in children are often different from adults. Similarly, children with epilepsy may present with comorbid symptoms that differ from children without epilepsy. The behavioral features of the seizures or the adverse effect of antiepileptic drugs (AEDs) may mask underlying comorbidities. AEDs are often attributed to behavior problems which often present even prior to the onset of seizures. This is challenging as it can lead to multiple medication changes for presumed behavioral side effects. When comorbid conditions are correctly diagnosed, there are concerns about exacerbation of the seizures, drug interactions and adverse effects when contemplating drug therapy. In addition, there is a paucity of data regarding drug efficacy for the comorbidities in childhood epilepsy.
2. Role of drug treatment of pediatric epilepsy in comorbidities
AEDs are the mainstay of treatment of childhood epilepsy. Although dietary therapy such as the ketogenic diet and surgical approaches are sometimes used in children with pharmacoresistant epilepsy, this group of patients makes up a relatively small percentage of cases.
It is important to recognize that a number of the first generation of AEDs (barbiturates [phenobarbital, primidone], benzodiazepines [clonazepam, diazepam, lorazepam], carbamazepine, ethosuximide, phenytoin and valproate) have adverse side effects that in some children could result in or exacerbate epilepsy comorbidities such as ADHD, depression, sleep disorders or anxiety (Table 2). While some of the second and third generation AEDs have fewer adverse effects, in any individual child the AED could contribute to the comorbidity. Children with the new onset of a comorbid condition following introduction of an AED should be re-evaluated to determine if the comorbidity is AED-related.
Table 2.
Antiepileptic drugs with favorable and unfavorable neuropsychological features [260].
| Drug | Favourable neuropsychological features |
Unfavourable neuropsychological features |
|---|---|---|
| Barbiturates | Anxiolytic | Depression Exacerbates ADHD |
| Benzodiazepines | Anxiolytic | Depression Exacerbates ADHD |
| Brivaracetam | None | None |
| Cannabinoids (CBD) | Anxiolytic | None |
| Carbamazepine | Mood stabilization (mild) | None |
| Eslicarbamazepine | None | None |
| Ethosuximide | None | None |
| Felbamate | None | Depression |
| Fenfluramine | Possible mood stabilization | None |
| Gabapentin | Anxiolytic | Irritability |
| Lamotrigine | Mood stabilization | None |
| Levetiracetam | None | Irritability Anxiety |
| Oxcarbazepine | Mood stabilization (mild) | None |
| Pregabalin | Anxiolytic | None |
| Rufinamide | None | None |
| Tiagabine | Anxiolytic | Depression Irritability |
| Topiramate | None | Depression Cognitive slowing |
| Zonisamide | None | Depression Cognitive slowing |
| Valproate | Mood stabilization | Irritability (uncommon) Agitation (uncommon) |
ADHD attention deficit hyperactive disorder
3. Mental Health Comorbidities
Mental health comorbidities have been recognized in people with epilepsy since the time of the ancient Greeks with Hippocrates opining in 400 B.C. “Melancholics ordinarily become epileptics, and epileptics, melancholics” [13, 14]. This statement remains true today, with a significant percentage of people with epilepsy having mental health conditions that remain undiagnosed and untreated. The term “mental health conditions” is used here to reflect a range of conditions that occur in children including depression, anxiety, ADHD, ASD, oppositional defiant disorder, conduct disorder, phobias, obsessive convulsive disease (OCD), panic attacks and functional neurological symptom disorders [15-19]. Many of the conditions listed are also comorbid with each other in children. For example, ASD, anxiety, depression and ADHD frequently co-occur (Fig. 2).
3.1. Depression and anxiety
Depressive and anxiety disorders are among the most common psychiatric comorbid conditions in epilepsy [2, 20-29]. Anxiety and depression are highly comorbid with each other, and together they are considered to belong to the broader category of internalizing disorders [30]. Since both conditions are comorbid with epilepsy [20], in this section they will be considered together.
Studies of the prevalence of common psychiatric comorbid conditions in pediatric epilepsy have estimated an overall risk of 21-60% for childhood psychopathology, a risk at least 3-6 fold higher than the general population [20-22], making the diagnosis of depression and anxiety in children with epilepsy difficult [27]. In children, depression does not necessarily equate to sadness and children with depression are more likely to show anxiety disorders, irritability, separation anxieties, and behavioral problems such as anger and aggression. The adolescent may present with academic decline, disruptive behavior, loss of interest in activities, problems with friends, aggression, irritability and suicidal ideation. Children with epilepsy and depression may also have psychogenic non-epileptic behavioral seizures [31, 32]. The clinical features of non-epileptic behavioral seizures are similar in children with epilepsy as those without epilepsy.
As noted by Hippocrates [13], epilepsy and depression are bidirectional with depression increasing the risk for epilepsy and epilepsy increasing the risk for depression. In both children and adults, a history of depression is associated with an increased risk for developing epilepsy [33-36]. Austin et al [10] reported that approximately a third of children with new-onset seizures had psychiatric issues, primarily depression and anxiety symptoms, prior to the onset of seizures.
In addition to being debilitating conditions, depression is associated with a worse prognosis for seizure control. In a study of 780 children and adults with epilepsy followed at a single center over a 20-year period, Hitiris et al.[37] found that psychiatric comorbidity, particularly depression, was correlated with pharmacoresistance. People with epilepsy and clinical depression reported higher levels of perceived seizure severity, recovery time and disability than did individuals with epilepsy without depression who experienced similar types of seizures [38]. These studies suggest that the deleterious neurobiological processes underpinning depression, anxiety and epilepsy interact to increase the extent of brain dysfunction and likelihood of developing medically intractable epilepsy.
In view of the detrimental effects of depression on quality of life and increased seizure susceptibility, identifying and treating depression is important. Unfortunately, until recently, antidepressants were thought to cause seizures and were contraindicated in children and adults with epilepsy. As recently as 2005, it was written that “most, if not all, antidepressant agents have a propensity to lower the seizure threshold, and most are associated with a risk for seizures” [39], making their use in people with a history of seizures problematic [40].
However, seizure risk in patients with epilepsy and depression who receive the newer generation antidepressants, particularly selective serotonin reuptake inhibitors (SSRI) and serotonin-norepinephrine reuptake inhibitor (SNRI) is low [39, 41, 42]. Kanner et al. [41] prospectively assessed the seizure frequency of 100 consecutive patients with partial and primary generalized epilepsy during a trial with sertraline, a SSRI, for the treatment of depression or OCD. While six patients (6%) experienced an increase in seizure frequency after starting sertraline, only one patient had a definite causal relationship between sertraline and seizure worsening. Thomé-Souza et al. [42] analyzed the impact of SSRIs on the severity and frequency of seizures in 36 children and adolescents with epilepsy and depression. All had an improvement in depression and seizures worsened in only two patients.
To extensively assess the effect of psychotropic medication on reported seizures, Alper et al. [43] examined data from 75,873 patients enrolled in Phase II and III clinical trials of psychotropic drugs that reported seizure incidence in United States trials between 1985 and 2004, The incidence of seizures among the active drug and placebo groups was compared with incidence data from the published rates of unprovoked seizures in the general population and from psychopharmacological clinical trials. The incidence of seizures was significantly lower among patients assigned to antidepressants, other than bupropion, compared to placebo (Table 3). The authors concluded that other than bupropion, second-generation antidepressants have an apparent anticonvulsant effect [43]. For anxiolytic drugs, alprazolam was shown to have a higher risk for seizures in the 1983 trial, but not in the 1990 trial.
Table 3.
Incidence of seizures in antidepressant and anti-anxiolytic drugs [43].
| Category | Incidence of Seizures | ||
|---|---|---|---|
| N | # of seizures | % | |
| Antidepressant drugs | |||
| Group 1 | |||
| Buproprion IR | 4419 | 26 | 6.0 |
| Group 2 | |||
| Citalopram | 4168 | 12 | 0.3 |
| Fluoxetine | 6000 | 12 | 0.2 |
| Venlafaxine | 2181 | 3 | 0.1 |
| Bupropion SR | 3094 | 3 | 0.1 |
| Paroxetine | 2963 | 2 | 0.07 |
| Nefazodone | 2256 | 1 | 0.04 |
| Mirtazapine | 2796 | 1 | 0.04 |
| Escitalopram | 715 | 0 | 0 |
| Duloxetine | 2314 | 0 | 0 |
| Sertraline | 2979 | 0 | 0 |
| Anti-anxiety drugs | |||
| Group 1 | |||
| Alprazolam (1983 trial) | 1723 | 46 | 2.7 |
| Group 2 | |||
| Alprazolam (1990 trial) | 1698 | 7 | 0.4 |
| Buspirone | 3558 | 3 | 0.8 |
Bupropion, an atypical antidepressant that acts as a norepinephrine–dopamine reuptake inhibitor and nicotinic receptor antagonist, is frequently associated with seizures [44, 45]. The incidence of seizures with bupropion is dose related [46]. Seizure risk was lower in clinical trials of sustained release (SR) versus immediate release (IR) formulation. Since peak concentrations are higher with IR versus SR formulations, the implication is that pharmacokinetic factors are involved in bupropion’s proconvulsive effect [47, 48]. Alprazolam is a short acting (half-life of ~11-13 hours) benzodiazepine used primarily in the treatment of anxiety disorders. While not all authors found an increased risk for seizures with alprazolam [49], when seizures do occur it is most commonly in the situation of sudden drug withdrawal [50, 51].
Thus, SSRIs and SNRIs now have a strong record of safety in children and are the most widely used group of drugs used to treat depression in epilepsy [52]. The relationship between L-5-hydroxytryptophan (5-HT), the immediate precursor of serotonin, and seizures [53-55] suggests serotonergic mechanisms are likely involved in the bidirectionality of epilepsy and depression. Agents that elevate extracellular serotonin levels, such as 5-HT and serotonin reuptake blockers, inhibit both focal and generalized seizures and depletion of brain 5-HT lowers the threshold to audiogenic, chemical and electrically evoked convulsions [56-60]. Genetic deletion of 5-HT2C receptors in mice causes audiogenic seizures [61] and knockout of 5-HT1A receptors reduces the threshold for kainate-induced seizures [62]. In humans, functional neuroimaging studies with positron emission tomography (PET) targeting the 5-HT1A receptor have suggested a potential common pathogenic role of 5-HT in epilepsy and depression [63-66]. Several AEDs including phenytoin, lamotrigine, carbamazepine, valproic acid and zonisamide cause an increase in extracellular 5-HT, and the elevated 5-HT is thought to contribute partially to their mechanism of action [55, 67-71]. In three small open trials using SSRIs in nondepressed adults with epilepsy, fluoxetine [72] and citralopam [73, 74] reduced seizure frequency. Fenfluramine, a potent 5-HT releaser/reuptake inhibitor activating multiple 5-HT receptor subtypes [75], is a highly efficacious AED in Dravet syndrome, a condition with severe epilepsy [76]. However, the antiepileptic effect of fenfluramine may not be linked fully to a serotonergic effect. Efficacy of fenfluramine may be related to its known modulatory effect at sigmal (σ1) receptors in vitro and in vivo [77].
While the evidence that SSRIs are safe in children with epilepsy is strong, the evidence that they are effective in treating depression is quite limited. In a Cochrane Library meta-analysis of eight studies (three randomized clinical trials and five prospective cohorts) investigating 471 children and adults with epilepsy treated for depression, treatment with a SSRI did not increase seizure frequency [78]. Evidence of the effectiveness of antidepressants in treating depressive symptoms associated with epilepsy, however, was very limited and only one small randomized clinical trial demonstrated a statistically significant effect of venlafaxine, a SNRI, on depressive symptoms. The authors concluded that high-quality evidence to inform the choice of antidepressant drug or class of drug in treating depression in people with epilepsy is not available [78].
Concomitant treatment of children with antidepressants or anxiolytic agents can result in drug interactions [79]. Most antidepressants inhibit one or more of the cytochrome P450 isoenzymes in the liver [80, 81]. Levels of enzyme-inducing AEDs (phenytoin, phenobarbital, primidone and carbamazepine) which induce the activity of CYP1A2, CYP2C9, CYP2C19 and CYP3A4, can increase with concomitant antidepressants. For example, elevated levels of carbamazepine have been observed with SSRI use [82-84]. Low doses of sertraline, venlafaxine, or citalopram have little effect on enzyme inhibition and therefore should not produce clinically significant interactions [80, 85]. Monitoring levels of AEDs can be helpful if toxicity is suspected in children receiving antidepressant or anxiolytic drugs.
In summary, children with epilepsy often have the comorbid conditions of depression and anxiety which can have profound effects on quality of life and possibly even seizure control. AEDs that can result in depression should be avoided, when possible (Table 1). While the new generation of antidepressants and anxiolytic agents are safe and may even reduce seizure burden, clinical evidence that antidepressants and anti-anxiolytics are effective in children with epilepsy is lacking.
3.2. ADHD
ADHD is a common comorbidity in childhood epilepsy with prevalence figures as high as 77% [86-93]. ADHD in epilepsy is associated with a significantly poorer quality of life with predominantly inattentiveness symptoms [94, 95]. In the general population, the prevalence of ADHD is approximately 5%, is more common in boys than girls, and most children have the combined type of ADHD with both attention deficits and hyperactive behavior. Children with epilepsy and ADHD differ from children without epilepsy and ADHD by having a higher proportion of inattentive symptoms and by an equal male to female ratio [86, 96].
ADHD has been found to have a bidirectional relationship with epilepsy, with either condition increasing the risk for the other [10, 11, 36, 96-98]. In studies of children with their first recognized, unprovoked seizure, ADHD was more frequent before the onset of the first seizure among children who developed epilepsy than among siblings without epilepsy or children with no additional seizures [10, 11]. A population based, case controlled study conducted among Icelandic children found that those with an unprovoked seizure were 2.5 times more likely than age- and gender-matched controls to have a prior history of ADHD [96]. In a study of 53 children aged 8 to 18 years with recent onset epilepsy (<1 year in duration) of idiopathic etiology, ADHD was two-fold higher in children with epilepsy than controls [36]. The increased prevalence of ADHD antedating epilepsy onset is consistent with the presence of underlying neurobiological factors independent of seizures and AEDs [36].
Approved drugs by the United States Food and Drug Administration (USFDA) for the treatment of ADHD include stimulants (amphetamine [AMP]–based and methylphenidate [MPH]–based agents) and nonstimulants (atomoxetine, clonidine, and guanfacine)(Table 4) [99, 100]. The primary pharmacologic effect of both amphetamine and methylphenidate is to increase central dopamine and norepinephrine activity, which impacts executive and attentional function [101]. Atomoxetine is a selective norepinephrine-reuptake inhibitor [102] and guanfacine and clonidine are selective α2-adrenergic agonists [103-105]. Stimulants are considered first-line therapy in children, adolescents, and adults with ADHD because of their greater efficacy over other approved drugs for ADHD [99, 100, 106, 107]
Table 4.
Drugs used to treat attention-deficit hyperactive disorder.
| Drug | Mechanism of Action | Drugs | Adverse Effects | Effect on Seizure Susceptibility |
|---|---|---|---|---|
| Methylphenidate | DA and NA reuptake inhibitor | Methylphenidate Dexmethylphenidate | Appetite suppression Emotional lability Insomnia Psychotic symptoms | None |
| D,L-Amphetamine | DA and NA reuptake inhibitor Monoamine oxidase activity inhibition VMAT-2 inhibition | Amphetamine sulfate Dextroamphetamine Lisdexamfetamine | Appetite suppression Emotional lability Insomnia Psychotic symptoms | None |
| Atomoxetine | NA-reuptake inhibition Increases DA indirectly | Atomoxetine | Appetite suppression Nausea Drowsiness | None |
| Guanfacine | Selective α2-adrenergic agonists | Guanfacine | Drowsiness Dizziness Dry mouth Constipation Tiredness | None |
| Clonidine | Selective α2-adrenergic agonists | Clonidine | Constipation. Dizziness Drowsiness Dry mouth Weakness Irritability | None |
DA dopaime; NA noradrenaline (norepinephrine)
AMP and MPH have been shown to exhibit comparable efficacy in two meta-analyses [108, 109], with other analyses reporting that AMP has moderately greater beneficial effects than MPH [110-112]. The tolerability and safety profiles of AMP and MPH in terms of adverse events, treatment discontinuation and cardiovascular effects are generally comparable [101].
As with antidepressants, treatment of ADHD in children with epilepsy has lagged behind the use of drugs in children without epilepsy because of the concern about stimulant medications lowering seizure threshold. In a study of 105 subjects with ADHD and epilepsy (mean age = 14.8±3.4 years), treatment with MPH resulted in aggravated seizures in 20% and worsening EEG abnormalities in 32% [113]. MPH was effective in controlling ADHD symptoms in both the seizure aggravation and non-aggravation groups. Children with poor seizure control or anxiety disorders at baseline were more likely to show an aggravation of seizures. The authors concluded that MPH may increase seizures and worsen EEG findings in individuals with ADHD and epilepsy [113]. In a randomized controlled trial of extended release MPH in 33 children (6-18 yrs) with ADHD and epilepsy taking AEDs participated in a multiple dose, double-blind placebo-controlled crossover trial with MPH [114]. MPH reduced ADHD symptoms more than placebo and there were too few seizures during the active and placebo arms to confidently assess seizure risk; however, considering exposure time, an increased daily risk of seizures with increasing dose of MPH was observed, suggesting potential safety concerns.
However, other studies did not find an exacerbation of seizures with MPH [93, 115, 116]. In a retrospective study of 36 children under the age of 18 with epilepsy and ADHD, symptoms were treated with MPH or an AMP preparation [116]. Responders were defined as those who remained on medication and had an improvement score of ≤ 2 on the Conners’ Global Index-Severity scale, a 7-point scale used to indicate severity of a patient’s symptoms with 1 indicating “normal” and 7 “extremely ill”. No patients who were seizure-free at the start of the medication trial experienced an increase in seizures. Of the patients having seizures at the start of the trial, one patient on MPH and two patients on AMP had increased seizures during the trial. Seizures returned to baseline frequency after stimulant discontinuation or anticonvulsant adjustment. MPH was associated with a higher response rate, with 12 of 19 given MPH compared with 4 of 17 given AMP responding.
To determine whether ADHD medication treatment increases risk of seizures among patients with and without preexisting seizures, Wiggs et al [93] assessed a database of 801,838 treated patients with ADHD to examine whether ADHD medication increased the likelihood of seizures among ADHD patients with and without a history of seizures. Patients with ADHD were at higher odds for any seizure compared with non-ADHD controls (odds ratio [OR] = 2.33, 95% confidence interval [CI] = 2.24–2.42 males; OR = 2.31, 95% CI = 2.22–2.42 females). In adjusted within-individual comparisons, ADHD medication was associated with lower odds of seizures among patients with (OR = 0.71, 95% CI = 0.60–0.85) and without (OR = 0.71, 95% CI = 0.62–0.82) prior seizures. Long-term within-individual comparisons suggested no evidence of an association between medication use and seizures among individuals with (OR = 0.87, 95% CI = 0.59–1.30) and without (OR = 1.01, 95% CI = 0.80–1.28) a seizure history. While the authors reaffirmed that patients with ADHD are at higher risk of seizures, ADHD medication was associated with lower risk of seizures in individuals while they were receiving medication.
In summary, children with epilepsy have a high prevalence of ADHD which can adversely affect school performance and reduce quality of life. ADHD often precedes epilepsy and is independent of seizures. In children with epilepsy and ADHD, a stepwise approach, beginning with non-drug interventions and then moving to pharmacological treatment in those most severely affected is warranted. Since in individual children some AEDs including phenobarbital, topiramate and valproate may worsen ADHD [88], AED therapy should be re-evaluated in children whose ADHD begins or is exacerbated with the introduction of an AED. If necessary, ADHD drug treatment can be safely started, and clinical response followed.
3.4. ASD
Autism, an early neurodevelopmental syndrome is characterized by abnormal language development, impairments in reciprocal social interactions, behavioral inflexibility and repetitive and ritualized behaviors [117]. While these are the core features of ASD, children with ASD often have co-morbid conditions such as ADHD, anxiety, depression, obsessive-compulsive disorder, schizophrenia and sleep disorders [118, 119].
There has been a known association of ASD with epilepsy for over fifty years [120]. The prevalence of epilepsy with ASD has varied widely, with a prevalence in some studies reaching almost 50% [121]. While population-based studies provide the best estimate of true prevalence of epilepsy in ASD. even those rates have been variable. In a prospective, population-based study of epilepsy in 120 young adults with autism, 38% had epilepsy [122]. In a study of 90 children with ASD between 6-12 years, 46% had epilepsy [123]. While there is a strong association between ASD and low IQ in children with epilepsy [123, 124], even in cases with a normal IQ, ASD is associated with an elevated risk of epilepsy [125-127].
Likewise, the prevalence of ASD in individuals with epilepsy is higher than in the general population [128, 129]. In a large population-based cohort of 64,188 people with epilepsy, the odds ratio of having ASD was 22.2 [130]. In a study of 48 young children (1-7 years) with epilepsy, 18% met ASD criteria and 40% met ADHD criteria [131]. Seizures early in life appear to place children at a particularly high risk for developing ASD [132-134].
In addition to an increased prevalence of epilepsy in individuals with ASD, there is also a marked increased incidence of epileptiform activity on EEGs in ASD. While interictal spikes occur in less than 5% of normal children without a history of epilepsy [135], studies have demonstrated that up to 60% of EEG records from individuals with autism have interictal spikes [136]. The high comorbidity rate between ASD and epilepsy and EEG epileptiform activity does not mean that there is a causal relationship between the conditions. More likely is the idea that epilepsy and ASD have a complex interaction and share common pathophysiological properties. This shared pathophysiology viewpoint is supported by the findings of Sundelin et al. [137] who found that ASD was more common in siblings and offspring of individuals with epilepsy than expected in the normal population. Seizure frequency is not associated with ASD [131, 138-140], suggesting that treating seizures does not prevent ASD from emerging.
The effectiveness of treatments for seizures has not been studied in individuals with ASD. Even though children with ASD and seizures appear to represent a large ASD subgroup, seizure treatments for individuals with ASD have not been systematically studied [141]. However, there is no evidence that seizures in children with ASD respond any differently to AEDs than seizures in children without ASD. Several lines of evidence point to valproate, lamotrigine, and levetiracetam as the most effective and tolerable AEDs for individuals with ASD [141]. A major consideration in the AED therapy of children with ASD is the side effect profile of the AED used. As discussed in Section 2, AEDs that exacerbate co-morbid conditions in ASD should be avoided.
There are currently no drugs that treat core autistic symptoms. Rather drug treatment aims at treating comorbid disorders, such as ADHD, sleep disturbances, irritability, agitation and aggressiveness and anxiety [142]. Psychomotor agitation or irritability is one of the more troublesome problems seen in children with ASD and atypical antipsychotics (also known as serotonin–dopamine antagonists) have emerged as helpful therapies.
The efficacy of risperidone, a dopamine type 2 (D2) and serotonin type 2A (5-HT2A) receptor antagonist, for the treatment of irritability in ASD, is supported by multiple placebo-controlled studies [143, 144]. In an 8-week double-blind, placebo-controlled trial of 101 children and adolescents with autism (ages 5–17 years), the Research Units on Pediatric Psychopharmacology (RUPP) Autism Network demonstrated a significant risperidone effect marked by a 69% drug treatment response versus a 12% response in those receiving placebo [143]. These findings were replicated by Shea et al. [145] who demonstrated in an 8 week double-blind, placebo-controlled trial of 79 children (5–12 years) with ASD that risperidone treatment was associated with significant improvement compared with placebo on several outcome measures [145].
Aripiprazole, a D2 and 5-HT1A receptor partial agonist and 5-HT2A receptor antagonist [146], has been shown to be effective in two double-blind, placebo-controlled studies, as well as in several open-label studies in the treatment of irritability in children with ASD [147, 148]. In an 8 week double-blind, placebo-controlled study involving 98 youth with ASD (ages 6–17 years), Owen et al. [148] reported a significant effect of aripiprazole on irritability, marked by a 52% drug treatment response rate compared with a 14% placebo response rate. Marcus et al. [149] reported on a fixed dose four arm (3 doses plus placebo) 8 week double-blind, placebo-controlled study of aripiprazole targeting irritability in children and adolescents with ASD (ages 6–17 years). Treatment response was seen in all three treatment arms compared with placebo.
Fallah et al [150] evaluated eight trials comparing four interventions (risperidone, aripiprazole, lurasidone, and placebo) in 878 children with ASD and irritability. Both risperidone and aripiprazole had significantly reduced Aberrant Behavioral Checklist Irritability (ABC-I) scores than placebo. Risperidone and aripiprazole are the only two medications approved by the US Food and Drug Administration (FDA) for the treatment of irritability associated with ASD. The risk of seizure with either risperidone or aripiprazole is quite low [151-153]. While there are no drug trials targeting irritability in children with epilepsy and ASD, there is no reason to believe the drugs would not be equal in efficacy, safety and tolerability in children with ASD and without epilepsy.
Cannabidiol is an FDA approved drug for the treatment of seizures associated with tuberous sclerosis complex, Lennox-Gastaut syndrome and Dravet syndrome. While it has been proposed as a drug that could be useful in treating the anxiety associated with ASD, there have been no conclusive studies indicating that any of the pathological symptoms of ASD such as hyperactivity, sleep disorders, self-injury, anxiety, behavioral problems, and communication benefit from the drug [154, 155].
4. Sleep Disorders
Sleep disorders and epilepsy are two conditions that interact with each other in a complex bi-directional way [156, 157]. Epilepsy can result in sleep disorders and sleep disorders can exacerbate epilepsy. There is a strong relationship between stages N1-N3 (drowsiness, light sleep and slow wave sleep [SWS]) and seizures and interictal EEG abnormalities [158]. Sleep is a powerful activator of seizures and interictal spikes, sharp waves and spike and wave complexes [158, 159]. The number of the epileptiform discharges as well as their propagation increase during all periods of sleep except during rapid-eye movement (REM) [160], unmasking the epileptic network that is often suppressed during wakefulness. Several epilepsy syndromes such as grand mal upon wakening [161], autosomal dominant nocturnal frontal lobe epilepsy [162] and benign Rolandic epilepsy (also termed Benign Epilepsy with Centro-Temporal Spikes) [163] have a strong relationship between seizures and sleep states.
Children with epilepsy are at risk for a variety of sleep disturbances including sleep anxiety, insomnia, fragmentation of sleep, excessive arousals, increased REM latency and reduced REM duration, obstructive sleep apnea and daytime sleepiness [19, 164-167]. Cortesi et al [166] assessed sleep problems in 89 children with idiopathic epilepsy compared with 49 siblings and 321 healthy control children using parental questionnaires. Children with epilepsy had poorer sleep quality and more sleep anxiety than the control population. In a study of 30 children with epilepsy, 80% exhibited sleep disruption because of either clinically significant obstructive sleep apnea syndrome, disturbance of sleep architecture or sleep fragmentation [164]. In a study of 79 school aged children with epilepsy by parental questionnaires, Stores et al [168] found a high incidence of poor-quality sleep and sleep anxieties which correlated with daytime behavioral abnormalities. Excessive daytime sleepiness occurs in a third to a half of patients with epilepsy [165]. although in many of the patients the sleepiness could be caused by sedating AEDs.
One of the most vexing sleep problems with children with epilepsy is insomnia which may be associated with cognitive, emotional, and psychosocial impairments resulting in significant caregiver burden [169]. Insomnia in children with epilepsy is complex and is often co-morbid with other conditions in children with epilepsy, including ADHD and anxiety [170]. Within the group of children with epilepsy, those with ASD are at particularly high risk for insomnia [119]. While children with epilepsy and other chronic illnesses go to bed at the same time as children without chronic illnesses, children with epilepsy have more problems falling asleep and more nighttime awakenings than healthy control children [171].
The consequences of insomnia in children are substantial, resulting in symptoms such as hyperactivity, inattention, learning difficulties, and impulsive behaviors [169, 172, 173]. In a study of 4175 children and adolescents aged 11-17 years, approximately 5% met criteria for chronic insomnia, and this was shown to increase risk for subsequent psychological problems, interpersonal difficulties and impairments in daily activities [174].
Seizures and interictal EEG activity can also have considerable effects on memory. SWS supports the consolidation of newly acquired information in memory. During SWS, slow oscillations in the delta activity (0-4 Hz), sleep spindles (11-15 Hz) and sharp wave-ripples (140–200 Hz) bandwidths [175-177] coordinate the re-activation and redistribution of hippocampus-dependent memories to neocortical sites, whereas REM sleep favors the subsequent synaptic consolidation of memories in the neocortex [178]. It known that both seizures and interictal spikes during sleep impact long-term memory consolidation [179] An association has been found between nocturnal interictal abnormalities and the frequency and severity of sleep disturbance [166].
Nocturnal seizures can disrupt sleep with both focal and generalized seizures resulting in arousal and disrupted sleep [180]. Some studies have reported that seizure frequency is positively associated with behavioral problems in children with epilepsy [166, 181, 182]. However, in a small series of 30 children with epilepsy, Becker et al. [164] found that behavioral problems were positively correlated with sleep disruption but not with seizure frequency, suggesting that behavioral problems may be a reflection of a coexisting sleep disorder in children with epilepsy. Further, children with epilepsy without nocturnal seizures can have sleep abnormalities, suggesting that sleep disturbances may be an intrinsic component in some patient with epilepsy [165].
AED therapy in children with epilepsy may play a role in sleep disturbances. As shown in Figure 3, a number of AEDs that have a GABAergic effect such as the benzodiazepines, barbiturates, vigabatrin and tiagabine can result in decreased sleep latency and daytime somnolence where others such as felbamate and lamotrigine can result in insomnia [183-187]. While AEDs can affect sleep efficiency and SWS and REM sleep durations (Figure 3), for the most part, these AED effects on sleep are not of clinical importance. While many of the AEDs are non-sedative, all of the AEDs can lead to sleepiness in selected patients, particularly if used at high doses. Finally, while AEDs have been shown to alter time spent asleep, sleep latency, and sleep architecture [187], sleep disturbances have been found to continue even after the discontinuation of anticonvulsant therapy [188], again suggesting that sleep disorders in children with epilepsy are intrinsic to the epilepsy, rather than caused by AEDs or seizures.
Figure 3.
Summary of the effects of antiepileptic drugs on sleep [183]. (↓ indicates decreases, ↔ indicates no changes, ↑ indicates increases, ? indicates effects unknown). REM rapid eye movement; SWS slow wave sleep.
Treatment of pediatric sleep disorders is likely to improve the quality of life for children with epilepsy by reducing seizure frequency as well as improving attentional difficulties [189]. For example, treatment of obstructive sleep apnea in patients with epilepsy may improve seizure control [189]. Parent-based sleep interventions are effective in randomized controlled trials for typically developing younger children, attention-deficit hyperactivity disorder and ASD [190-193] and applicable for children with epilepsy. If behavioral strategies are not helpful and contributing coexisting conditions and use of concomitant medications have been addressed, melatonin can be useful [194] }.
5. Cognitive Dysfunction
Cognitive dysfunction is a major concern for children with epilepsy. In a study of children with intellectual disabilities, approximately 15% developed epilepsy by 22 years of age [195], reflecting a 43-fold increased risk in comparison to children without intellectual disability [196]. When adjustment is made for age, socioeconomic status and gender, among children with intellectual disabilities, a 9-fold increased risk to have one or more seizures was found when compared to controls [197]. Furthermore, the presence of cerebral palsy associated with intellectual disability strongly increases the risk for developing epilepsy [195, 198]. Executive dysfunction, such as deficits in working memory and planning abilities, has been noted in many children and adolescents with epilepsy [199, 200].
The distribution of IQ scores of children with epilepsy is skewed toward lower values [201, 202] and the number of children experiencing difficulties in school because of learning disabilities or behavioral problems is greater than in the normal population [97, 203, 204]. Even children with normal IQs and well-controlled seizures are at high risk for learning problems [203]. Memory impairment, mental slowing and attentional deficits are the most frequently reported problems.
A variety of factors - many of which are not intrinsically associated with having seizures or treatment - impact the neurobehavioral status of people with epilepsy [205]. For example, a cohort of people over the age of 15 years with newly diagnosed epilepsy and without other neurological disorders had significantly worse results than healthy volunteers in several cognitive domains even before AEDs were started [206]. Similarly, Hermann and colleagues [207] found that children with new-onset epilepsy had cognitive impairment and academic underachievement in comparison to children without epilepsy.
While most of the cognitive problems that occur in children precede the onset of epilepsy, young children with active epilepsy have a significant risk for a variety of problems involving cognition and behavior. Children appear to be more vulnerable to the adverse effects of seizures, EEG abnormalities, and epilepsy treatment than adults [208-211]. There is evidence that some children with epilepsy are slow in their cognitive development [202] or even have progressive declines of IQ on serial intelligence tests and behavioral and psychiatric deterioration over time [201, 210-215]. In a community-based cohort of 198 children, aged <8 years with new-onset epilepsy followed prospectively and reassessed with the Wechsler Intelligence Scales for Children (WISC-III), 8-9 years later, Berg et al. [216] found that children with medically intractable seizures had an 11.4 point lower full scale IQ with similar decrements in each WISC-III domain. The authors concluded that young children with pharmacoresistant seizures suffered from cognitive decline over time.
For children who have cognitive problems before the onset of the epilepsy, there is little one can do to improve cognition with pharmacological interventions. However, if the child has another comorbid condition such as depression or ADHD, pharmacological treatment could be helpful in allowing the child to maximize cognition. If the child has cognitive decline following the onset of the seizures, optimizing seizure control with AEDs, preferably using monotherapy, may be helpful. While all AEDs are likely to have some effect on cognition based on their mechanism of action on neural networks, for the most part, AEDs do not have major adverse effects on cognition in children with epilepsy [217-219]. In the first generation of AEDs (carbamazepine, clonazepam, ethosuximide, felbamate, gabapentin, phenobarbital, phenytoin, primidone and valproate), cognitive issues have been raised with clonazepam, phenobarbital and primidone [220, 221]. Children treated with phenobarbital for febrile seizures have had cognitive issues compared to non-treated children [222, 223]. In a review of the second and third generation AEDs (eslicarbazepine, lacosamide, lamotrigine, levetiracetam, oxcarbazepine, perampanel, rufinamide, tiagabine, topiramate, vigabatrin and zonisamide), it was found that the drugs had few cognitive side effects [219]. The highest rate of cognitive side effects occurred with topiramate which has been associated with cognitive impairment (attention, memory, and language function) in children in some [224, 225], but not all studies [226].
There is one important group of children with epileptic encephalopathies in which treatment of seizures and interictal EEG abnormalities can influence cognitive outcome. Epileptic encephalopathies refer to a group of disorders in which the unremitting epileptic activity contributes to severe cognitive and behavioral impairments above and beyond what might be expected from the underlying pathology alone, and these can worsen over time, leading to progressive cerebral dysfunction [227]. The epileptic encephalopathies encompass disorders such as early myoclonic encephalopathy, Ohtahara syndrome, migrating partial seizures of infancy, infantile spasms and Lennox-Gastaut syndrome. Inherent in the concept of epileptic encephalopathy is the idea that suppression of epileptic activity may improve cognition and behavior. Effective and early intervention may in fact improve seizure control and developmental outcome in some cases [228-230]. In two other rare syndromes in the epileptic encephalopathies category, Landau-Kleffner Syndrome [231-233] and Electrical Status Epilepticus of Sleep (ESES)(also known as Continuous Spike Wave of Sleep [CSWS]) [234-236], there is evidence that interictal discharges are contributing to the neurological symptoms. However, there is little evidence that treating interictal spikes improves cognition [237].
In conclusion, cognitive impairment is a common comorbidity in childhood epilepsy and usually precedes or occurs concomitantly with the onset of epilepsy. In most cases the etiology of the epilepsy is the cause of the cognitive impairment. Recurrent seizures and interictal spikes, or both, account for cognitive decline in only a small percentage of children. In this group of children, optimizing seizure control with AEDs that do not contribute to the cognitive impairment is important.
5. Migraine
Migraine and epilepsy are common comorbidities in children [238-240] and contribute to a poor quality of life [241, 242]. In children, an association between migraine and several epilepsy syndromes - childhood epilepsy with occipital paroxysms, Benign Rolandic Epilepsy and Panayiotopoulos syndrome have been reported [243, 244]. Migraines have been each associated with a more severe epilepsy course [242]. In a study of adults with epilepsy with and without migraine, patients with epilepsy and migraine had a significantly lower cumulative probability of being seizure-free over 10 years compared with patients without migraine [245].
As with many of the other comorbid conditions associated with childhood epilepsy, there is a bidirectional relationship between migraine and epilepsy, where having a history of one condition is associated with an increased risk for the other [246, 247]. Migraine is associated with a fourfold increased risk for developing epilepsy, an association explained by migraine with aura (odds ratio, 8.1; 95% confidence interval, 2.7-24.3), whereas migraine without aura did not increase risk for epilepsy in children [246]. Velioğlu and colleagues [248] found that people (>15 years) with both epilepsy and migraine had poorer seizure control than people with epilepsy but without migraine. The nature of the association of migraine and epilepsy is complex but suggests that a common pathophysiological mechanism may be present.
A diverse group of medications is used to prevent migraine attacks including antiepileptics, antidepressants, antihistamines, and antihypertensive agents, in addition to newer classes of medications; yet, there remains a dearth of controlled studies in children [249]. There have also been no formal, prospective trials designed to treat migraine in children with epilepsy. Indeed, for pediatric migraine in children without epilepsy, clinical practice guidelines for migraine treatment are consensus based rather than evidence based with no USFDA–approved migraine prevention medication for children younger than 12 years of age [250]. In a randomized, double-blind, placebo-controlled trial of amitriptyline, topiramate and placebo in 361 children and adolescents 8 to 17 years of age with migraine, there were no significant differences in reduction in headache frequency or headache-related disability in childhood and adolescent migraine with amitriptyline, topiramate, or placebo over a period of 24 weeks and a higher rate of adverse events in the patients receiving active drug [250].
Two AEDs, topiramate and valproate, are approved for migraine prevention in children (≥ 12 yrs) and adults by the USFDA [251, 252]. Topiramate is the most widely used migraine preventative agent and is approved for children [243, 253, 254]. Valproate is also widely used and is effective in adolescent and adult patients with migraine [254]. Efficacy of topiramate and valproate in children with epilepsy and migraine is not known.
Available evidence does not allow robust conclusions regarding the efficacy of AEDs other than topiramate and valproate in migraine [255]. In a Cochran review the pooled evidence derived from trials of gabapentin suggests that it is not efficacious for the prophylaxis of episodic migraine in adults and is associated with significant side effects [255]. There is no published evidence from controlled trials of pregabalin for the prophylaxis of episodic migraine in adults [255]. Acetazolamide, carbamazepine, clonazepam, lamotrigine, oxcarbazepine, and vigabatrin were not found to be more effective than placebo in reducing headache frequency [256]. In small trials, carbamazepine and levetiracetam were better than placebo in reducing headache [256].
For the acute therapy of migraines in children with epilepsy, ibuprofen, prochlorperazine, and certain triptan medications are the most effective and safe agents for acute management of migraine and other benign headache disorders in the pediatric population [257, 258]. These drugs are not contraindicated in children with epilepsy. Of note, sumatriptan has been effectively used in post-ictal migraine headaches [259].
In summary, children with epilepsy have a higher than expected increase in migraine, which can adversely affect quality of life and potentially contribute to seizure severity. In the child with both migraine and epilepsy, consideration may be given to using an AED which has proven to be efficacious for migraine in children (>12 years) and adults, namely, topiramate and valproate. However, there is limited data indicating that topiramate, valproate or any other drugs are effective in preventing migraine in children with epilepsy. Abortive therapy used for migraines in children without epilepsy can be used safely in children with epilepsy.
6. Conclusions
Comorbidities in children with epilepsy are common and can greatly add to the disease burden of the condition. Recognizing that certain AEDs can cause or exacerbate the comorbidities can lead to a better therapeutic plan for the child. When the comorbid condition can be treated with an AED this should be considered. Most drugs used to treat depression, anxiety, ADHD and migraine in children without epilepsy can be used safely in children with epilepsy. While there are a paucity of studies providing clear data on efficacy of drugs to treat the comorbidities of childhood epilepsy, there is no evidence that drugs used to treat comorbidities in children without epilepsy are any less effective in children with epilepsy.
KEY POINTS.
Children with epilepsy have a high prevalence of comorbid conditions.
For most comorbid conditions epilepsy is not causative but has a bidirectional association with the comorbidity.
Most psychotropic medications are not contraindicated in children with epilepsy.
Drug treatment of comorbid conditions in children with epilepsy is similar to treatment of children without comorbidities.
Efficacy information on treatment of comorbid conditions in childhood epilepsy is based on limited data.
Acknowledgments
Funding:
This work was supported by the NIH (NINDS) Grants NS108765; NS108296.
Footnotes
Conflicts of interest/Competing interests:
None
Ethics approval:
Nor applicable
Consent to participate:
Not applicable
Consent for publication:
Not applicable
Availability of data and material:
All articles cited are available in PubMed
Code availability:
Not applicable
Reference List
- 1.Berg AT. Epilepsy, cognition, and behavior: The clinical picture. Epilepsia. 2011. January;52 Suppl 1(Suppl 1):7–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tellez-Zenteno JF, Patten SB, Jetté N, Williams J, Wiebe S. Psychiatric comorbidity in epilepsy: a population-based analysis. Epilepsia. 2007. December;48(12):2336–44. [DOI] [PubMed] [Google Scholar]
- 3.Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014. April/2014;55(4):475–82. [DOI] [PubMed] [Google Scholar]
- 4.Feinstein AR. The Pre-Therapeutic Classification of Co-Morbidity in Chronic Disease. J Chronic Dis. 1970. December;23(7):455–68. [DOI] [PubMed] [Google Scholar]
- 5.Welton JM, Walker C, Riney K, Ng A, Todd L, D'Souza WJ. Quality of life and its association with comorbidities and adverse events from antiepileptic medications: Online survey of patients with epilepsy in Australia. Epilepsy Behav. 2020. March;104(Pt A):106856. [DOI] [PubMed] [Google Scholar]
- 6.Morningstar M, Hung A, Mattson WI, Gedela S, Ostendorf AP, Nelson EE. Internalizing symptoms in intractable pediatric epilepsy: Structural and functional brain correlates. Epilepsy Behav. 2020. February;103(Pt A):106845. [DOI] [PubMed] [Google Scholar]
- 7.Riechmann J, Willems LM, Boor R, Kieslich M, Knake S, Langner C, et al. Quality of life and correlating factors in children, adolescents with epilepsy, and their caregivers: A cross-sectional multicenter study from Germany. Seizure. 2019. July;69:92–8. [DOI] [PubMed] [Google Scholar]
- 8.Puka K, Ferro MA, Camfield CS, Levin SD, Smith ML, Wiebe S, et al. Trajectories of quality of life 10 years following a diagnosis of epilepsy in childhood. Epilepsia. 2020. July;61(7):1453–63. [DOI] [PubMed] [Google Scholar]
- 9.Institute of Medicine. EpilepsyAcross the Spectrum: Promoting Health and Understanding: The National Academies Press; 2012. [PubMed] [Google Scholar]
- 10.Austin JK, Harezlak J, Dunn DW, Huster GA, Rose DF, Ambrosius WT. Behavior problems in children before first recognized seizures. Pediatrics. 2001. January/2001;107(1):115–22. [DOI] [PubMed] [Google Scholar]
- 11.Dunn DW, Austin JK, Huster GA. Behaviour problems in children with new-onset epilepsy. Seizure. 1997. August/1997;6(4):283–7. [PubMed] [Google Scholar]
- 12.Kanner AM. Psychiatric comorbidity in patients with developmental disorders and epilepsy: a practical approach to its diagnosis and treatment. Epilepsy Behav. 2002. December;3(6s1):7–13. [DOI] [PubMed] [Google Scholar]
- 13.Lewis A Melancholia: a historical review. . J Mental Sci 1934;80:1. [Google Scholar]
- 14.Temkin O The Falling Sickness. A History of Epilepsy From the Greeks to th Begining of Modern Neurology. Baltimore: Johns Hopkins Press; 1971. [Google Scholar]
- 15.Swinkels WA, Kuyk J, van Dyck R, Spinhoven P. Psychiatric comorbidity in epilepsy. Epilepsy Behav. 2005. August;7(1):37–50. [DOI] [PubMed] [Google Scholar]
- 16.Dunn DW, Austin JK, Perkins SM. Prevalence of psychopathology in childhood epilepsy: categorical and dimensional measures. Dev Med Child Neurol. 2009. May/2009;51(5):364–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Salayev KA, Sanne B, Salayev R. Psychiatric and Behavioural Problems in Children and Adolescents with Epilepsy. East Asian Arch Psychiatry. 2017. September;27(3):106–14. [PubMed] [Google Scholar]
- 18.Gaitatzis A, Carroll K, Majeed A, J WS. The epidemiology of the comorbidity of epilepsy in the general population. Epilepsia. 2004. December;45(12):1613–22. [DOI] [PubMed] [Google Scholar]
- 19.Gaitatzis A, Trimble MR, Sander JW. The psychiatric comorbidity of epilepsy. Acta Neurol Scand. 2004. October;110(4):207–20. [DOI] [PubMed] [Google Scholar]
- 20.Ekinci O, Titus JB, Rodopman AA, Berkem M, Trevathan E. Depression and anxiety in children and adolescents with epilepsy: prevalence, risk factors, and treatment. Epilepsy Behav. 2009. January/2009;14(1):8–18. [DOI] [PubMed] [Google Scholar]
- 21.Ott D, Caplan R, Guthrie D, Siddarth P, Komo S, Shields WD, et al. Measures of psychopathology in children with complex partial seizures and primary generalized epilepsy with absence. J Am Acad Child Adolesc Psychiatry. 2001. August;40(8):907–14. [DOI] [PubMed] [Google Scholar]
- 22.Ott D, Siddarth P, Gurbani S, Koh S, Tournay A, Shields WD, et al. Behavioral disorders in pediatric epilepsy: unmet psychiatric need. Epilepsia. 2003. April;44(4):591–7. [DOI] [PubMed] [Google Scholar]
- 23.Plioplys S Depression in children and adolescents with epilepsy. Epilepsy Behav. 2003. October;4 Suppl 3:S39–45. [DOI] [PubMed] [Google Scholar]
- 24.Piazzini A, Canevini MP, Maggiori G, Canger R. Depression and Anxiety in Patients with Epilepsy. Epilepsy Behav. 2001. October;2(5):481–9. [DOI] [PubMed] [Google Scholar]
- 25.Ettinger AB, Weisbrot DM, Nolan EE, Gadow KD, Vitale SA, Andriola MR, et al. Symptoms of depression and anxiety in pediatric epilepsy patients. Epilepsia. 1998. June;39(6):595–9. [DOI] [PubMed] [Google Scholar]
- 26.Williams J, Steel C, Sharp GB, DelosReyes E, Phillips T, Bates S, et al. Anxiety in children with epilepsy. Epilepsy Behav. 2003. December;4(6):729–32. [DOI] [PubMed] [Google Scholar]
- 27.Caplan R, Siddarth P, Gurbani S, Hanson R, Sankar R, Shields WD. Depression and anxiety disorders in pediatric epilepsy. Epilepsia. 2005. May/2005;46(5):720–30. [DOI] [PubMed] [Google Scholar]
- 28.Reilly C, Agnew R, Neville BG. Depression and anxiety in childhood epilepsy: a review. Seizure. 2011. October/2011;20(8):589–97. [DOI] [PubMed] [Google Scholar]
- 29.Yang C, Hao Z, Mao Y, Xu Q, Zhao L, Zhang LL. Depression in children with epilepsy from western China: A cross-sectional survey. Medicine (Baltimore). 2020. June 12;99(24):e20647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kalin NH. The Critical Relationship Between Anxiety and Depression. Am J Psychiatry. 2020. May 1;177(5):365–7. [DOI] [PubMed] [Google Scholar]
- 31.Tamer SK. The pediatric non-epileptic seizure. Indian J Pediatr. 1997. Sep-Oct;64(5):671–6. [DOI] [PubMed] [Google Scholar]
- 32.Verrotti A, Agostinelli S, Mohn A, Manco R, Coppola G, Franzoni E, et al. Clinical features of psychogenic non-epileptic seizures in prepubertal and pubertal patients with idiopathic epilepsy. Neurol Sci. 2009. August;30(4):319–23. [DOI] [PubMed] [Google Scholar]
- 33.Forsgren L, Nystrom L. An incident case-referent study of epileptic seizures in adults. Epilepsy Res. 1990. May/1990;6(1):66–81. [DOI] [PubMed] [Google Scholar]
- 34.Hesdorffer DC, Hauser WA, Annegers JF, Cascino G. Major depression is a risk factor for seizures in older adults. Ann Neurol. 2000. February/2000;47(2):246–9. [PubMed] [Google Scholar]
- 35.Hesdorffer DC, Hauser WA, Olafsson E, Ludvigsson P, Kjartansson O. Depression and suicide attempt as risk factors for incident unprovoked seizures. Ann Neurol. 2006. January/2006;59(1):35–41. [DOI] [PubMed] [Google Scholar]
- 36.Jones JE, Watson R, Sheth R, Caplan R, Koehn M, Seidenberg M, et al. Psychiatric comorbidity in children with new onset epilepsy. Dev Med Child Neurol. 2007. July;49(7):493–7. [DOI] [PubMed] [Google Scholar]
- 37.Hitiris N, Mohanraj R, Norrie J, Sills GJ, Brodie MJ. Predictors of pharmacoresistant epilepsy. Epilepsy Res. 2007. July;75(2-3):192–6. [DOI] [PubMed] [Google Scholar]
- 38.Cramer JA, Blum D, Reed M, Fanning K. The influence of comorbid depression on seizure severity. Epilepsia. 2003. December;44(12):1578–84. [DOI] [PubMed] [Google Scholar]
- 39.Montgomery SA. Antidepressants and seizures: emphasis on newer agents and clinical implications. Int J Clin Pract. 2005. December;59(12):1435–40. [DOI] [PubMed] [Google Scholar]
- 40.Rosenstein DL, Nelson JC, Jacobs SC. Seizures associated with antidepressants: a review. J Clin Psychiatry. 1993. August;54(8):289–99. [PubMed] [Google Scholar]
- 41.Kanner AM, Kozak AM, Frey M. The Use of Sertraline in Patients with Epilepsy: Is It Safe? Epilepsy Behav. 2000. April;1(2):100–5. [DOI] [PubMed] [Google Scholar]
- 42.Thomé-Souza MS, Kuczynski E, Valente KD. Sertraline and fluoxetine: safe treatments for children and adolescents with epilepsy and depression. Epilepsy Behav. 2007. May;10(3):417–25. [DOI] [PubMed] [Google Scholar]
- 43.Alper K, Schwartz KA, Kolts RL, Khan A. Seizure incidence in psychopharmacological clinical trials: an analysis of Food and Drug Administration (FDA) summary basis of approval reports. Biol Psychiatry. 2007. August 15;62(4):345–54. [DOI] [PubMed] [Google Scholar]
- 44.Pesola GR, Avasarala J. Bupropion seizure proportion among new-onset generalized seizures and drug related seizures presenting to an emergency department. J Emerg Med. 2002. April;22(3):235–9. [DOI] [PubMed] [Google Scholar]
- 45.Górska N, Słupski J, Cubała WJ, Wiglusz MS, Gałuszko-Węgielnik M. Antidepressants in epilepsy. Neurol Neurochir Pol. 2018. Nov-Dec;52(6):657–61. [DOI] [PubMed] [Google Scholar]
- 46.Davidson J Seizures and bupropion: a review. J Clin Psychiatry. 1989. July;50(7):256–61. [PubMed] [Google Scholar]
- 47.Dunner DL, Zisook S, Billow AA, Batey SR, Johnston JA, Ascher JA. A prospective safety surveillance study for bupropion sustained-release in the treatment of depression. J Clin Psychiatry. 1998. July;59(7):366–73. [DOI] [PubMed] [Google Scholar]
- 48.Jefferson JW, Pradko JF, Muir KT. Bupropion for major depressive disorder: Pharmacokinetic and formulation considerations. Clin Ther. 2005. November;27(11):1685–95. [DOI] [PubMed] [Google Scholar]
- 49.Jonas JM, Cohon MS. A comparison of the safety and efficacy of alprazolam versus other agents in the treatment of anxiety, panic, and depression: a review of the literature. J Clin Psychiatry. 1993. October;54 Suppl:25–45; discussion 6-8. [PubMed] [Google Scholar]
- 50.Noyes R Jr., Perry PJ, Crowe RR, Coryell WH, Clancy J, Yamada T, et al. Seizures following the withdrawal of alprazolam. J Nerv Ment Dis. 1986. January;174(1):50–2. [DOI] [PubMed] [Google Scholar]
- 51.Naylor MW, Grunhaus L, Cameron O. Myoclonic seizures after abrupt withdrawal from phenelzine and alprazolam. J Nerv Ment Dis. 1987. February;175(2):111–4. [DOI] [PubMed] [Google Scholar]
- 52.Dunn DW. Should pediatric neurologists play a role in the management of the most common psychiatric comorbidities in children with epilepsy? Practical considerations. Epilepsy Behav. 2019. September;98(Pt B):314–7. [DOI] [PubMed] [Google Scholar]
- 53.Richerson GB, Buchanan GF. The serotonin axis: Shared mechanisms in seizures, depression, and SUDEP. Epilepsia. 2011. January;52 Suppl 1(Suppl 1):28–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kanner AM. Depression and epilepsy: A bidirectional relation? Epilepsia. 2011. January;52 Suppl 1:21–7. [DOI] [PubMed] [Google Scholar]
- 55.Bagdy G, Kecskemeti V, Riba P, Jakus R. Serotonin and epilepsy. J Neurochem. 2007. February;100(4):857–73. [DOI] [PubMed] [Google Scholar]
- 56.Kilian M, Frey HH. Central monoamines and convulsine thresholds in mice and rats. Neuropharmacology. 1973. July;12(7):681–92. [DOI] [PubMed] [Google Scholar]
- 57.Buterbaugh GG. Effect of drugs modifying central serotonergic function on the response of extensor and nonextensor rats to maximal electroshock. Life Sci. 1978. December 11;23(24):2393–404. [DOI] [PubMed] [Google Scholar]
- 58.Browning RA, Hoffmann WE, Simonton RL. Changes in seizure susceptibility after intracerebral treatment with 5,7-dihydroxytryptamine: role of serotonergic neurons. Annals of the New York Academy of Sciences. 1978. June 12;305:437–56. [DOI] [PubMed] [Google Scholar]
- 59.Statnick MA, Maring-Smith ML, Clough RW, Wang C, Dailey JW, Jobe PC, et al. Effect of 5,7-dihydroxytryptamine on audiogenic seizures in genetically epilepsy-prone rats. Life Sci. 1996;59(21):1763–71. [DOI] [PubMed] [Google Scholar]
- 60.Yan QS, Jobe PC, Cheong JH, Ko KH, Dailey JW. Role of serotonin in the anticonvulsant effect of fluoxetine in genetically epilepsy-prone rats. Naunyn Schmiedebergs Arch Pharmacol. 1994. August;350(2):149–52. [DOI] [PubMed] [Google Scholar]
- 61.Brennan TJ, Seeley WW, Kilgard M, Schreiner CE, Tecott LH. Sound-induced seizures in serotonin 5-HT2c receptor mutant mice. Nat Genet. 1997. August;16(4):387–90. [DOI] [PubMed] [Google Scholar]
- 62.Sarnyai Z, Sibille EL, Pavlides C, Fenster RJ, McEwen BS, Toth M. Impaired hippocampal-dependent learning and functional abnormalities in the hippocampus in mice lacking serotonin(1A) receptors. Proceedings of the National Academy of Sciences of the United States of America. 2000. December 19;97(26):14731–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Toczek MT, Carson RE, Lang L, Ma Y, Spanaki MV, Der MG, et al. PET imaging of 5-HT1A receptor binding in patients with temporal lobe epilepsy. Neurology. 2003. March 11;60(5):749–56. [DOI] [PubMed] [Google Scholar]
- 64.Giovacchini G, Toczek MT, Bonwetsch R, Bagic A, Lang L, Fraser C, et al. 5-HT 1A receptors are reduced in temporal lobe epilepsy after partial-volume correction. J Nucl Med. 2005. July;46(7):1128–35. [PMC free article] [PubMed] [Google Scholar]
- 65.Merlet I, Ostrowsky K, Costes N, Ryvlin P, Isnard J, Faillenot I, et al. 5-HT1A receptor binding and intracerebral activity in temporal lobe epilepsy: an [18F]MPPF-PET study. Brain. 2004. April;127(Pt 4):900–13. [DOI] [PubMed] [Google Scholar]
- 66.Theodore WH. Does Serotonin Play a Role in Epilepsy? Epilepsy Curr. 2003. September;3(5):173–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Isaac M. Serotonergic 5-HT2C receptors as a potential therapeutic target for the design antiepileptic drugs. Curr Top Med Chem. 2005;5(1):59–67. [DOI] [PubMed] [Google Scholar]
- 68.Aad G, Abbott B, Abdallah J, Abdelalim AA, Abdesselam A, Abdinov O, et al. Search for new particles in two-jet final states in 7 TeV proton-proton collisions with the ATLAS detector at the LHC. Phys Rev Lett. 2010. October/15/2010;105(16):161801. [DOI] [PubMed] [Google Scholar]
- 69.Dailey JW, Reith ME, Yan QS, Li MY, Jobe PC. Carbamazepine increases extracellular serotonin concentration: lack of antagonism by tetrodotoxin or zero Ca2+. Eur J Pharmacol. 1997. June 11;328(2-3):153–62. [DOI] [PubMed] [Google Scholar]
- 70.Okada M, Kaneko S, Hirano T, Ishida M, Kondo T, Otani K, et al. Effects of zonisamide on extracellular levels of monoamine and its metabolite, and on Ca2+ dependent dopamine release. Epilepsy Res. 1992. November;13(2):113–9. [DOI] [PubMed] [Google Scholar]
- 71.Bonnycastle DD, Giarman NJ, Paasonen MK. Anticonvulsant compounds and 5-hydroxytryptamine in rat brain. Br J Pharmacol Chemother. 1957. June;12(2):228–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Favale E, Rubino V, Mainardi P, Lunardi G, Albano C. Anticonvulsant effect of fluoxetine in humans. Neurology. 1995. October;45(10):1926–7. [DOI] [PubMed] [Google Scholar]
- 73.Favale E, Audenino D, Cocito L, Albano C. The anticonvulsant effect of citalopram as an indirect evidence of serotonergic impairment in human epileptogenesis. Seizure. 2003. July;12(5):316–8. [DOI] [PubMed] [Google Scholar]
- 74.Specchio LM, Iudice A, Specchio N, La Neve A, Spinelli A, Galli R, et al. Citalopram as treatment of depression in patients with epilepsy. Clin Neuropharmacol. 2004. May-Jun;27(3):133–6. [DOI] [PubMed] [Google Scholar]
- 75.Sourbron J, Schneider H, Kecskés A, Liu Y, Buening EM, Lagae L, et al. Serotonergic Modulation as Effective Treatment for Dravet Syndrome in a Zebrafish Mutant Model. ACS Chem Neurosci. 2016. May 18;7(5):588–98. [DOI] [PubMed] [Google Scholar]
- 76.Lagae L, Sullivan J, Knupp K, Laux L, Polster T, Nikanorova M, et al. Fenfluramine hydrochloride for the treatment of seizures in Dravet syndrome: a randomised, double-blind, placebo-controlled trial. Lancet. 2020. December 21;394(10216):2243–54. [DOI] [PubMed] [Google Scholar]
- 77.Martin P, de Witte PAM, Maurice T, Gammaitoni A, Farfel G, Galer B. Fenfluramine acts as a positive modulator of sigma-1 receptors. Epilepsy Behav. 2020. April;105:106989. [DOI] [PubMed] [Google Scholar]
- 78.Maguire MJ, Weston J, Singh J, Marson AG. Antidepressants for people with epilepsy and depression. Cochrane Database Syst Rev. 2014. December 3;2014(12):Cd010682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bosak M, Słowik A, Iwańska A, Lipińska M, Turaj W. Co-medication and potential drug interactions among patients with epilepsy. Seizure. 2019. March;66:47–52. [DOI] [PubMed] [Google Scholar]
- 80.Nemeroff CB, DeVane CL, Pollock BG. Newer antidepressants and the cytochrome P450 system. Am J Psychiatry. 1996. March;153(3):311–20. [DOI] [PubMed] [Google Scholar]
- 81.Harden CL. The co-morbidity of depression and epilepsy: epidemiology, etiology, and treatment. Neurology. 2002. September 24;59(6 Suppl 4):S48–55. [DOI] [PubMed] [Google Scholar]
- 82.Grimsley SR, Jann MW, Carter JG, D'Mello AP, D'Souza MJ. Increased carbamazepine plasma concentrations after fluoxetine coadministration. Clin Pharmacol Ther. 1991. July;50(1):10–5. [DOI] [PubMed] [Google Scholar]
- 83.Martinelli V, Bocchetta A, Palmas AM, Del Zompo M. An interaction between carbamazepine and fluvoxamine. Br J Clin Pharmacol. 1993. December;36(6):615–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ashton AK, Wolin RE. Nefazodone-induced carbamazepine toxicity. Am J Psychiatry. 1996. May;153(5):733. [DOI] [PubMed] [Google Scholar]
- 85.Ereshefsky L Drug-drug interactions involving antidepressants: focus on venlafaxine. J Clin Psychopharmacol. 1996. June;16(3 Suppl 2):37S–50S; discussion S-3S. [DOI] [PubMed] [Google Scholar]
- 86.Dunn DW, Austin JK, Harezlak J, Ambrosius WT. ADHD and epilepsy in childhood. Dev Med Child Neurol. 2003. January;45(1):50–4. [PubMed] [Google Scholar]
- 87.Cohen R, Senecky Y, Shuper A, Inbar D, Chodick G, Shalev V, et al. Prevalence of epilepsy and attention-deficit hyperactivity (ADHD) disorder: a population-based study. J Child Neurol. 2013. January;28(1):120–3. [DOI] [PubMed] [Google Scholar]
- 88.Verrotti A, Moavero R, Panzarino G, Di Paolantonio C, Rizzo R, Curatolo P. The Challenge of Pharmacotherapy in Children and Adolescents with Epilepsy-ADHD Comorbidity. Clin Drug Investig. 2018. January;38(1):1–8. [DOI] [PubMed] [Google Scholar]
- 89.Parisi P, Moavero R, Verrotti A, Curatolo P. Attention deficit hyperactivity disorder in children with epilepsy. Brain Dev. 2010. January;32(1):10–6. [DOI] [PubMed] [Google Scholar]
- 90.Fastenau PS, Jianzhao S, Dunn DW, Austin JK. Academic underachievement among children with epilepsy: proportion exceeding psychometric criteria for learning disability and associated risk factors. J Learn Disabil. 2008. May-Jun;41(3):195–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Brikell I, Ghirardi L, D'Onofrio BM, Dunn DW, Almqvist C, Dalsgaard S, et al. Familial Liability to Epilepsy and Attention-Deficit/Hyperactivity Disorder: A Nationwide Cohort Study. Biol Psychiatry. 2018. January 15;83(2):173–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Chou IC, Chang YT, Chin ZN, Muo CH, Sung FC, Kuo HT, et al. Correlation between epilepsy and attention deficit hyperactivity disorder: a population-based cohort study. PloS one. 2013;8(3):e57926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wiggs KK, Chang Z, Quinn PD, Hur K, Gibbons R, Dunn D, et al. Attention-deficit/hyperactivity disorder medication and seizures. Neurology. 2018. March 27;90(13):e1104–e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ekinci O, Okuyaz Ç, Erdoğan S, Gunes S, Ekinci N, Kalınlı M, et al. Attention-Deficit Hyperactivity Disorder (ADHD) in Epilepsy and Primary ADHD: Differences in Symptom Dimensions and Quality of Life. J Child Neurol. 2017. December;32(14):1083–91. [DOI] [PubMed] [Google Scholar]
- 95.Alibas H, Koytak PK, Ekinci G, Uluc K. A case with leukoencephalopathy with brainstem and spinal cord involvement and elevated lactate (LBSL) with Its Characteristic Clinical and Neuroimaging Findings. Clin Neuroradiol. 2014. September;24(3):297–300. [DOI] [PubMed] [Google Scholar]
- 96.Hesdorffer DC, Ludvigsson P, Olafsson E, Gudmundsson G, Kjartansson O, Hauser WA. ADHD as a risk factor for incident unprovoked seizures and epilepsy in children. Arch Gen Psychiatry. 2004. July/2004;61(7):731–6. [DOI] [PubMed] [Google Scholar]
- 97.Williams J, Griebel ML, Dykman RA. Neuropsychological patterns in pediatric epilepsy. Seizure. 1998 1998;7:223–8. [DOI] [PubMed] [Google Scholar]
- 98.Holtmann M, Becker K, Kentner-Figura B, Schmidt MH. Increased frequency of rolandic spikes in ADHD children. Epilepsia. 2003. September;44(9):1241–4. [DOI] [PubMed] [Google Scholar]
- 99.Atkinson M, Hollis C. NICE guideline: attention deficit hyperactivity disorder. Arch Dis Child Educ Pract Ed. 2010. February;95(1):24–7. [DOI] [PubMed] [Google Scholar]
- 100.Wolraich M, Brown L, Brown RT, DuPaul G, Earls M, Feldman HM, et al. ADHD: clinical practice guideline for the diagnosis, evaluation, and treatment of attention-deficit/hyperactivity disorder in children and adolescents. Pediatrics. 2011. November;128(5):1007–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Faraone SV. The pharmacology of amphetamine and methylphenidate: Relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neuroscience and biobehavioral reviews. 2018. April;87:255–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Reed VA, Buitelaar JK, Anand E, Day KA, Treuer T, Upadhyaya HP, et al. The Safety of Atomoxetine for the Treatment of Children and Adolescents with Attention-Deficit/Hyperactivity Disorder: A Comprehensive Review of Over a Decade of Research. CNS Drugs. 2016. July;30(7):603–28. [DOI] [PubMed] [Google Scholar]
- 103.Biederman J, Melmed RD, Patel A, McBurnett K, Konow J, Lyne A, et al. A randomized, double-blind, placebo-controlled study of guanfacine extended release in children and adolescents with attention-deficit/hyperactivity disorder. Pediatrics. 2008. January;121(1):e73–84. [DOI] [PubMed] [Google Scholar]
- 104.Sallee FR, Lyne A, Wigal T, McGough JJ. Long-term safety and efficacy of guanfacine extended release in children and adolescents with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol. 2009. June;19(3):215–26. [DOI] [PubMed] [Google Scholar]
- 105.Jain R, Segal S, Kollins SH, Khayrallah M. Clonidine extended-release tablets for pediatric patients with attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2011. February;50(2):171–9. [DOI] [PubMed] [Google Scholar]
- 106.Bolea-Alamañac B, Nutt DJ, Adamou M, Asherson P, Bazire S, Coghill D, et al. Evidence-based guidelines for the pharmacological management of attention deficit hyperactivity disorder: update on recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2014. March;28(3):179–203. [DOI] [PubMed] [Google Scholar]
- 107.Gorman DA, Gardner DM, Murphy AL, Feldman M, Bélanger SA, Steele MM, et al. Canadian guidelines on pharmacotherapy for disruptive and aggressive behaviour in children and adolescents with attention-deficit hyperactivity disorder, oppositional defiant disorder, or conduct disorder. Can J Psychiatry. 2015. February;60(2):62–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Faraone SV, Glatt SJ. A comparison of the efficacy of medications for adult attention-deficit/hyperactivity disorder using meta-analysis of effect sizes. J Clin Psychiatry. 2010. June;71(6):754–63. [DOI] [PubMed] [Google Scholar]
- 109.Catalá-López F, Hutton B, Núñez-Beltrán A, Page MJ, Ridao M, Macías Saint-Gerons D, et al. The pharmacological and non-pharmacological treatment of attention deficit hyperactivity disorder in children and adolescents: A systematic review with network meta-analyses of randomised trials. PloS one. 2017;12(7):e0180355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Faraone SV, Buitelaar J. Comparing the efficacy of stimulants for ADHD in children and adolescents using meta-analysis. Eur Child Adolesc Psychiatry. 2010. April;19(4):353–64. [DOI] [PubMed] [Google Scholar]
- 111.Joseph A, Ayyagari R, Xie M, Cai S, Xie J, Huss M, et al. Comparative efficacy and safety of attention-deficit/hyperactivity disorder pharmacotherapies, including guanfacine extended release: a mixed treatment comparison. Eur Child Adolesc Psychiatry. 2017. August;26(8):875–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Stuhec M, Munda B, Svab V, Locatelli I. Comparative efficacy and acceptability of atomoxetine, lisdexamfetamine, bupropion and methylphenidate in treatment of attention deficit hyperactivity disorder in children and adolescents: a meta-analysis with focus on bupropion. J Affect Disord. 2015. June 1;178:149–59. [DOI] [PubMed] [Google Scholar]
- 113.Park J, Choi HW, Yum MS, Ko TS, Shon SH, Kim HW. Relationship Between Aggravation of Seizures and Methylphenidate Treatment in Subjects with Attention-Deficit/Hyperactivity Disorder and Epilepsy. J Child Adolesc Psychopharmacol. 2018. October;28(8):537–46. [DOI] [PubMed] [Google Scholar]
- 114.Gonzalez-Heydrich J, Whitney J, Waber D, Forbes P, Hsin O, Faraone SV, et al. Adaptive phase I study of OROS methylphenidate treatment of attention deficit hyperactivity disorder with epilepsy. Epilepsy Behav. 2010. July;18(3):229–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Verotti A, Trotta D, Cutarella R, Pascarella R, Morgese G, Chiarelli F. Effects of antiepileptic drugs on evoked potentials in epileptic children. Pediatr Neurol. 2000. 2000;23:397–402. [DOI] [PubMed] [Google Scholar]
- 116.Gonzalez-Heydrich J, Hsin O, Gumlak S, Kimball K, Rober A, Azeem MW, et al. Comparing stimulant effects in youth with ADHD symptoms and epilepsy. Epilepsy Behav. 2014. July;36:102–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Schumann CM, Bloss CS, Barnes CC, Wideman GM, Carper RA, Akshoomoff N, et al. Longitudinal magnetic resonance imaging study of cortical development through early childhood in autism. J Neurosci. 2010. March/24/2010;30(12):4419–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Howes OD, Rogdaki M, Findon JL, Wichers RH, Charman T, King BH, et al. Autism spectrum disorder: Consensus guidelines on assessment, treatment and research from the British Association for Psychopharmacology. J Psychopharmacol. 2018. January;32(1):3–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ballester P, Richdale AL, Baker EK, Peiró AM. Sleep in autism: A biomolecular approach to aetiology and treatment. Sleep Med Rev. 2020. July 9;54:101357. [DOI] [PubMed] [Google Scholar]
- 120.Kanner L Autistic disturbances of affective contact. Acta Paedopsychiatr. 1968. 1968;35(4):100–36. [PubMed] [Google Scholar]
- 121.Spence SJ, Schneider MT. The role of epilepsy and epileptiform EEGs in autism spectrum disorders. Pediatr Res. 2009. June/2009;65(6):599–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Danielsson S, Gillberg IC, Billstedt E, Gillberg C, Olsson I. Epilepsy in young adults with autism: a prospective population-based follow-up study of 120 individuals diagnosed in childhood. Epilepsia. 2005. June/2005;46(6):918–23. [DOI] [PubMed] [Google Scholar]
- 123.Anukirthiga B, Mishra D, Pandey S, Juneja M, Sharma N. Prevalence of Epilepsy and Inter-Ictal Epileptiform Discharges in Children with Autism and Attention-Deficit Hyperactivity Disorder. Indian J Pediatr. 2019. October;86(10):897–902. [DOI] [PubMed] [Google Scholar]
- 124.Viscidi EW, Triche EW, Pescosolido MF, McLean RL, Joseph RM, Spence SJ, et al. Clinical characteristics of children with autism spectrum disorder and co-occurring epilepsy. PLoS ONE. 2013. 2013;8(7):e67797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Amiet C, Gourfinkel-An I, Bouzamondo A, Tordjman S, Baulac M, Lechat P, et al. Epilepsy in autism is associated with intellectual disability and gender: evidence from a meta-analysis. Biol Psychiatry. 2008. October/1/2008;64(7):577–82. [DOI] [PubMed] [Google Scholar]
- 126.Olsson I, Steffenburg S, Gillberg C. Epilepsy in autism and autisticlike conditions. A population-based study. Arch Neurol. 1988. June/1988;45(6):666–8. [DOI] [PubMed] [Google Scholar]
- 127.Elia M, Musumeci SA, Ferri R, Bergonzi P. Clinical and neurophysiological aspects of epilepsy in subjects with autism and mental retardation. Am J Ment Retard. 1995. July/1995;100(1):6–16. [PubMed] [Google Scholar]
- 128.Woolfenden S, Sarkozy V, Ridley G, Coory M, Williams K. A systematic review of two outcomes in autism spectrum disorder - epilepsy and mortality. Dev Med Child Neurol. 2012. April/2012;54(4):306–12. [DOI] [PubMed] [Google Scholar]
- 129.Tuchman R, Cuccaro M. Epilepsy and autism: neurodevelopmental perspective. Curr Neurol Neurosci Rep. 2011. August/2011;11(4):428–34. [DOI] [PubMed] [Google Scholar]
- 130.Selassie AW, Wilson DA, Martz GU, Smith GG, Wagner JL, Wannamaker BB. Epilepsy beyond seizure: a population-based study of comorbidities. Epilepsy Res. 2014. February/2014;108(2):305–15. [DOI] [PubMed] [Google Scholar]
- 131.Reilly C, Atkinson P, Memon A, Jones C, Dabydeen L, Helen Cross J, et al. Autism, ADHD and parent-reported behavioural difficulties in young children with epilepsy. Seizure. 2019. October;71:233–9. [DOI] [PubMed] [Google Scholar]
- 132.Saemundsen E, Ludvigsson P, Hilmarsdottir I, Rafnsson V. Autism spectrum disorders in children with seizures in the first year of life - a population-based study. Epilepsia. 2007. September/2007;48(9):1724–30. [DOI] [PubMed] [Google Scholar]
- 133.Saemundsen E, Ludvigsson P, Rafnsson V. Risk of autism spectrum disorders after infantile spasms: a population-based study nested in a cohort with seizures in the first year of life. Epilepsia. 2008. November/2008;49(11):1865–70. [DOI] [PubMed] [Google Scholar]
- 134.Saemundsen E, Magnusson P, Georgsdottir I, Egilsson E, Rafnsson V. Prevalence of autism spectrum disorders in an Icelandic birth cohort. BMJ Open. 2013. 2013;3(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Eeg-Olofsson O, Petersén I, Selldén U. The development of the electroencephalogram in normal children from the age of 1 through 15 years: Paroxysmal activity. Neuropädiatrie. 1971. 1971;2:375–404. [DOI] [PubMed] [Google Scholar]
- 136.Hughes JR, Melyn M. EEG and seizures in autistic children and adolescents: further findings with therapeutic implications. Clin EEG Neurosci. 2005. January/2005;36(1):15–20. [DOI] [PubMed] [Google Scholar]
- 137.Sundelin HE, Larsson H, Lichtenstein P, Almqvist C, Hultman CM, Tomson T, et al. Autism and epilepsy: A population-based nationwide cohort study. Neurology. 2016. July 12;87(2):192–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Reilly C, Atkinson P, Das KB, Chin RF, Aylett SE, Burch V, et al. Neurobehavioral comorbidities in children with active epilepsy: a population-based study. Pediatrics. 2014. June/2014;133(6):e1586–e93. [DOI] [PubMed] [Google Scholar]
- 139.Austin JK, Caplan R. Behavioral and psychiatric comorbidities in pediatric epilepsy: toward an integrative model. Epilepsia. 2007. September;48(9):1639–51. [DOI] [PubMed] [Google Scholar]
- 140.Plioplys S, Dunn DW, Caplan R. 10-year research update review: psychiatric problems in children with epilepsy. J Am Acad Child Adolesc Psychiatry. 2007. November;46(11):1389–402. [DOI] [PubMed] [Google Scholar]
- 141.Frye RE, Rossignol D, Casanova MF, Brown GL, Martin V, Edelson S, et al. A review of traditional and novel treatments for seizures in autism spectrum disorder: findings from a systematic review and expert panel. Front Public Health. 2013. September 13;1:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lamy M, Pedapati EV, Dominick KL, Wink LK, Erickson CA. Recent Advances in the Pharmacological Management of Behavioral Disturbances Associated with Autism Spectrum Disorder in Children and Adolescents. Paediatr Drugs. 2020. October;22(5):473–83. [DOI] [PubMed] [Google Scholar]
- 143.McCracken JT, McGough J, Shah B, Cronin P, Hong D, Aman MG, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med. 2002. August 1;347(5):314–21. [DOI] [PubMed] [Google Scholar]
- 144.Erickson CA, Posey DJ, Stigler KA, McDougle CJ. Pharmacologic treatment of autism and related disorders. Pediatr Ann. 2007. September;36(9):575–85. [DOI] [PubMed] [Google Scholar]
- 145.Shea S, Turgay A, Carroll A, Schulz M, Orlik H, Smith I, et al. Risperidone in the treatment of disruptive behavioral symptoms in children with autistic and other pervasive developmental disorders. Pediatrics. 2004. November;114(5):e634–41. [DOI] [PubMed] [Google Scholar]
- 146.Potkin SG, Saha AR, Kujawa MJ, Carson WH, Ali M, Stock E, et al. Aripiprazole, an antipsychotic with a novel mechanism of action, and risperidone vs placebo in patients with schizophrenia and schizoaffective disorder. Arch Gen Psychiatry. 2003. July;60(7):681–90. [DOI] [PubMed] [Google Scholar]
- 147.Marcus RN, Owen R, Kamen L, Manos G, McQuade RD, Carson WH, et al. A placebo-controlled, fixed-dose study of aripiprazole in children and adolescents with irritability associated with autistic disorder. J Am Acad Child Adolesc Psychiatry. 2009. November;48(11):1110–9. [DOI] [PubMed] [Google Scholar]
- 148.Owen R, Sikich L, Marcus RN, Corey-Lisle P, Manos G, McQuade RD, et al. Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics. 2009. December;124(6):1533–40. [DOI] [PubMed] [Google Scholar]
- 149.Marcus RN, Owen R, Manos G, Mankoski R, Kamen L, McQuade RD, et al. Aripiprazole in the treatment of irritability in pediatric patients (aged 6-17 years) with autistic disorder: results from a 52-week, open-label study. J Child Adolesc Psychopharmacol. 2011. June;21(3):229–36. [DOI] [PubMed] [Google Scholar]
- 150.Fallah MS, Shaikh MR, Neupane B, Rusiecki D, Bennett TA, Beyene J. Atypical Antipsychotics for Irritability in Pediatric Autism: A Systematic Review and Network Meta-Analysis. J Child Adolesc Psychopharmacol. 2019. April;29(3):168–80. [DOI] [PubMed] [Google Scholar]
- 151.Holzhausen SP, Guerreiro MM, Baccin CE, Montenegro MA. Use of risperidone in children with epilepsy. Epilepsy Behav. 2007. May;10(3):412–6. [DOI] [PubMed] [Google Scholar]
- 152.Górska N, Słupski J, Cubała WJ. Antipsychotic drugs in epilepsy. Neurol Neurochir Pol. 2019;53(6):408–12. [DOI] [PubMed] [Google Scholar]
- 153.Wu CS, Wang SC, Yeh IJ, Liu SK. Comparative risk of seizure with use of first- and second-generation antipsychotics in patients with schizophrenia and mood disorders. J Clin Psychiatry. 2016. May;77(5):e573–9. [DOI] [PubMed] [Google Scholar]
- 154.Premoli M, Aria F, Bonini SA, Maccarinelli G, Gianoncelli A, Pina SD, et al. Cannabidiol: Recent advances and new insights for neuropsychiatric disorders treatment. Life Sci. 2019. May 1;224:120–7. [DOI] [PubMed] [Google Scholar]
- 155.Agarwal R, Burke SL, Maddux M. Current state of evidence of cannabis utilization for treatment of autism spectrum disorders. BMC Psychiatry. 2019. October 29;19(1):328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Nobili L, de Weerd A, Rubboli G, Beniczky S, Derry C, Eriksson S, et al. Standard procedures for the diagnostic pathway of sleep-related epilepsies and comorbid sleep disorders: A European Academy of Neurology, European Sleep Research Society and International League against Epilepsy-Europe consensus review. J Sleep Res. 2020. September 21:e13184. [DOI] [PubMed] [Google Scholar]
- 157.Wirrell E, Blackman M, Barlow K, Mah J, Hamiwka L. Sleep disturbances in children with epilepsy compared with their nearest-aged siblings. Dev Med Child Neurol. 2005. November;47(11):754–9. [DOI] [PubMed] [Google Scholar]
- 158.Halász P, Bódizs R, Ujma PP, Fabó D, Szűcs A. Strong relationship between NREM sleep, epilepsy and plastic functions - A conceptual review on the neurophysiology background. Epilepsy Res. 2019. February;150:95–105. [DOI] [PubMed] [Google Scholar]
- 159.Kotagal P, Yardi N. The relationship between sleep and epilepsy. Seminars in pediatric neurology. 2008. June;15(2):42–9. [DOI] [PubMed] [Google Scholar]
- 160.Frauscher B, von Ellenrieder N, Dubeau F, Gotman J. EEG desynchronization during phasic REM sleep suppresses interictal epileptic activity in humans. Epilepsia. 2016. June;57(6):879–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Janz D Epilepsy with grand mal on awakening and sleep-waking cycle. Clin Neurophysiol. 2000. September;111 Suppl 2:S103–10. [DOI] [PubMed] [Google Scholar]
- 162.Menghi V, Bisulli F, Tinuper P, Nobili L. Sleep-related hypermotor epilepsy: prevalence, impact and management strategies. Nat Sci Sleep. 2018;10:317–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Aricò M, Arigliani E, Giannotti F, Romani M. ADHD and ADHD-related neural networks in benign epilepsy with centrotemporal spikes: A systematic review. Epilepsy Behav. 2020. September 21;112:107448. [DOI] [PubMed] [Google Scholar]
- 164.Becker DA, Fennell EB, Carney PR. Daytime behavior and sleep disturbance in childhood epilepsy. Epilepsy Behav. 2004. October;5(5):708–15. [DOI] [PubMed] [Google Scholar]
- 165.Foldvary-Schaefer N, Grigg-Damberger M. Sleep and epilepsy: what we know, don't know, and need to know. J Clin Neurophysiol. 2006. February;23(1):4–20. [DOI] [PubMed] [Google Scholar]
- 166.Cortesi F, Giannotti F, Ottaviano S. Sleep problems and daytime behavior in childhood idiopathic epilepsy. Epilepsia. 1999. November;40(11):1557–65. [DOI] [PubMed] [Google Scholar]
- 167.Becker DA, Fennell EB, Carney PR. Sleep disturbance in children with epilepsy. Epilepsy Behav. 2003. December;4(6):651–8. [DOI] [PubMed] [Google Scholar]
- 168.Stores G, Wiggs L, Campling G. Sleep disorders and their relationship to psychological disturbance in children with epilepsy. Child Care Health Dev. 1998. January;24(1):5–19. [DOI] [PubMed] [Google Scholar]
- 169.Owens JA, Moturi S. Pharmacologic treatment of pediatric insomnia. Child Adolesc Psychiatr Clin N Am. 2009. October;18(4):1001–16. [DOI] [PubMed] [Google Scholar]
- 170.Brown KM, Malow BA. Pediatric Insomnia. Chest. 2016. May;149(5):1332–9. [DOI] [PubMed] [Google Scholar]
- 171.Hysing M, Sivertsen B, Stormark KM, Elgen I, Lundervold AJ. Sleep in children with chronic illness, and the relation to emotional and behavioral problems--a population-based study. J Pediatr Psychol. 2009. July;34(6):665–70. [DOI] [PubMed] [Google Scholar]
- 172.Cortese S, Konofal E, Yateman N, Mouren MC, Lecendreux M. Sleep and alertness in children with attention-deficit/hyperactivity disorder: a systematic review of the literature. Sleep. 2006. April;29(4):504–11. [PubMed] [Google Scholar]
- 173.Cortese S, Konofal E, Lecendreux M, Arnulf I, Mouren MC, Darra F, et al. Restless legs syndrome and attention-deficit/hyperactivity disorder: a review of the literature. Sleep. 2005. August 1;28(8):1007–13. [DOI] [PubMed] [Google Scholar]
- 174.Roberts RE, Roberts CR, Duong HT. Chronic insomnia and its negative consequences for health and functioning of adolescents: a 12-month prospective study. J Adolesc Health. 2008. March;42(3):294–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Ramadan W, Eschenko O, Sara SJ. Hippocampal sharp wave/ripples during sleep for consolidation of associative memory. PloS one. 2009. August 20;4(8):e6697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Farmer CA, Chilakamarri P, Thurm AE, Swedo SE, Holmes GL, Buckley AW. Spindle activity in young children with autism, developmental delay, or typical development. Neurology. 2018. July 10;91(2):e112–e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Oyanedel CN, Durán E, Niethard N, Inostroza M, Born J. Temporal associations between sleep slow oscillations, spindles and ripples. Eur J Neurosci. 2020. July 11. [DOI] [PubMed] [Google Scholar]
- 178.Diekelmann S, Born J. The memory function of sleep. Nature reviews Neuroscience. 2010. February;11(2):114–26. [DOI] [PubMed] [Google Scholar]
- 179.Lambert I, Tramoni-Negre E, Lagarde S, Roehri N, Giusiano B, Trebuchon-Da Fonseca A, et al. Hippocampal Interictal Spikes during Sleep Impact Long-Term Memory Consolidation. Ann Neurol. 2020. June;87(6):976–87. [DOI] [PubMed] [Google Scholar]
- 180.Bazil CW, Castro LH, Walczak TS. Reduction of rapid eye movement sleep by diurnal and nocturnal seizures in temporal lobe epilepsy. Arch Neurol. 2000. March/2000;57(3):363–8. [DOI] [PubMed] [Google Scholar]
- 181.Austin JK, Risinger MW, Beckett LA. Correlates of behavior problems in children with epilepsy. Epilepsia. 1992. Nov-Dec;33(6):1115–22. [DOI] [PubMed] [Google Scholar]
- 182.Hoare P The development of psychiatric disorder among schoolchildren with epilepsy. Dev Med Child Neurol. 1984. February/1984;26(1):3–13. [DOI] [PubMed] [Google Scholar]
- 183.Bazil CW. Effects of antiepileptic drugs on sleep structure : are all drugs equal? CNS Drugs. 2003;17(10):719–28. [DOI] [PubMed] [Google Scholar]
- 184.Jain SV, Glauser TA. Effects of epilepsy treatments on sleep architecture and daytime sleepiness: an evidence-based review of objective sleep metrics. Epilepsia. 2014. January;55(1):26–37. [DOI] [PubMed] [Google Scholar]
- 185.Cardenas JF, Rho JM, Ng YT. Reversible lamotrigine-induced neurobehavioral disturbances in children with epilepsy. J Child Neurol. 2010. February;25(2):182–7. [DOI] [PubMed] [Google Scholar]
- 186.Byars AW, Byars KC, Johnson CS, DeGrauw TJ, Fastenau PS, Perkins S, et al. The relationship between sleep problems and neuropsychological functioning in children with first recognized seizures. Epilepsy Behav. 2008. November;13(4):607–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Foldvary N. Sleep and Epilepsy. Curr Treat Options Neurol. 2002. March;4(2):129–35. [DOI] [PubMed] [Google Scholar]
- 188.Hoeppner JB, Garron DC, Cartwright RD. Self-reported sleep disorder symptoms in epilepsy. Epilepsia. 1984. August;25(4):434–7. [DOI] [PubMed] [Google Scholar]
- 189.Malow BA, Weatherwax KJ, Chervin RD, Hoban TF, Marzec ML, Martin C, et al. Identification and treatment of obstructive sleep apnea in adults and children with epilepsy: a prospective pilot study. Sleep Med. 2003. November;4(6):509–15. [DOI] [PubMed] [Google Scholar]
- 190.Hiscock H, Sciberras E, Mensah F, Gerner B, Efron D, Khano S, et al. Impact of a behavioural sleep intervention on symptoms and sleep in children with attention deficit hyperactivity disorder, and parental mental health: randomised controlled trial. Bmj. 2015. January 20;350:h68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Sciberras E, Mulraney M, Mensah F, Oberklaid F, Efron D, Hiscock H. Sustained impact of a sleep intervention and moderators of treatment outcome for children with ADHD: a randomised controlled trial. Psychol Med. 2020. January;50(2):210–9. [DOI] [PubMed] [Google Scholar]
- 192.Johnson CR, Turner KS, Foldes E, Brooks MM, Kronk R, Wiggs L. Behavioral parent training to address sleep disturbances in young children with autism spectrum disorder: a pilot trial. Sleep Med. 2013. October;14(10):995–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Mindell JA, Kuhn B, Lewin DS, Meltzer LJ, Sadeh A. Behavioral treatment of bedtime problems and night wakings in infants and young children. Sleep. 2006. October;29(10):1263–76. [PubMed] [Google Scholar]
- 194.Williams Buckley A, Hirtz D, Oskoui M, Armstrong MJ, Batra A, Bridgemohan C, et al. Practice guideline: Treatment for insomnia and disrupted sleep behavior in children and adolescents with autism spectrum disorder: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology. 2020. March 3;94(9):392–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Goulden KJ, Shinnar S, Koller H, Katz M, Richardson SA. Epilepsy in children with mental retardation: a cohort study. Epilepsia. 1991. Sep-Oct;32(5):690–7. [DOI] [PubMed] [Google Scholar]
- 196.Hauser WA, Annegers JF, Kurland LT. Incidence of epilepsy and unprovoked seizures in Rochester, Minnesota: 1935-1984. Epilepsia. 1993. May/1993;34(3):453–68. [DOI] [PubMed] [Google Scholar]
- 197.Richardson SA, Koller H, Katz M, McLaren J. Seizures and epilepsy in a mentally retarded population over the first 22 years of life. Appl Res Ment Retard. 1980;1(1-2):123–38. [DOI] [PubMed] [Google Scholar]
- 198.Carlsson M, Hagberg G, Olsson I. Clinical and aetiological aspects of epilepsy in children with cerebral palsy. Dev Med Child Neurol. 2003. June;45(6):371–6. [DOI] [PubMed] [Google Scholar]
- 199.MacAllister WS, Bender HA, Whitman L, Welsh A, Keller S, Granader Y, et al. Assessment of executive functioning in childhood epilepsy: the Tower of London and BRIEF. Child Neuropsychol. 2012;18(4):404–15. [DOI] [PubMed] [Google Scholar]
- 200.MacAllister WS, Vasserman M, Coulehan K, Hall AF, Bender HA. Cognitive estimations as a measure of executive dysfunction in childhood epilepsy. Child Neuropsychol. 2016;22(1):65–80. [DOI] [PubMed] [Google Scholar]
- 201.Farwell JR, Dodrill CB, Batzel LW. Neuropsychological abilities of children with epilepsy. Epilepsia. 1985. 1985;26:395–400. [DOI] [PubMed] [Google Scholar]
- 202.Neyens LG, Aldenkamp AP, Meinardi HM. Prospective follow-up of intellectual development in children with a recent onset of epilepsy. Epilepsy Res. 1999. 1999;34:85–90. [DOI] [PubMed] [Google Scholar]
- 203.Bailet LL, Turk WR. The impact of childhood epilepsy on neurocognitive and behavioral performance: a prospective longitudinal study. Epilepsia. 2000. April/2000;41(4):426–31. [DOI] [PubMed] [Google Scholar]
- 204.Sillanpaa M, Jalava M, Kaleva O, Shinnar S. Long-term prognosis of seizures with onset in childhood. N Engl J Med. 1998. June/11/1998;338(24):1715–22. [DOI] [PubMed] [Google Scholar]
- 205.Hermann B, Seidenberg M. Epilepsy and cognition. Epilepsy Curr. 2007. Jan-Feb;7(1):1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Taylor J, Kolamunnage-Dona R, Marson AG, Smith PE, Aldenkamp AP, Baker GA. Patients with epilepsy: cognitively compromised before the start of antiepileptic drug treatment? Epilepsia. 2010. January;51(1):48–56. [DOI] [PubMed] [Google Scholar]
- 207.Hermann BP, Seidenberg M, Dow C, Jones J, Rutecki P, Bhattacharya A, et al. Cognitive prognosis in chronic temporal lobe epilepsy. Ann Neurol. 2006. July/2006;60(1):80–7. [DOI] [PubMed] [Google Scholar]
- 208.Bjornaes H, Stabell K, Henriksen O, Loyning Y. The effects of refractory epilepsy on intellectual functioning in children and adults. A longitudinal study. Seizure. 2001. June/2001;10(4):250–9. [DOI] [PubMed] [Google Scholar]
- 209.Holmes GL. Cognitive impairment in epilepsy: the role of network abnormalities. Epileptic Disord. 2015. June/2015;17(2):101–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Berg AT, Smith SN, Frobish D, Beckerman B, Levy SR, Testa FM, et al. Longitudinal assessment of adaptive behavior in infants and young children with newly diagnosed epilepsy: influences of etiology, syndrome, and seizure control. Pediatrics. 2004. September;114(3):645–50. [DOI] [PubMed] [Google Scholar]
- 211.Berg AT, Zelko FA, Levy SR, Testa FM. Age at onset of epilepsy, pharmacoresistance, and cognitive outcomes: a prospective cohort study. Neurology. 2012. September 25;79(13):1384–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Tromp SC, Weber JW, Aldenkamp AP, Arends J, vander L, I, Diepman L. Relative influence of epileptic seizures and of epilepsy syndrome on cognitive function. J Child Neurol. 2003. June/2003;18(6):407–12. [DOI] [PubMed] [Google Scholar]
- 213.Bourgeois BFD, Prensky AL, Palkes HS, Talent BK, Busch SG. Intelligence in epilepsy: A prospective study in children. Ann Neurol. 1983. 1983;14:438–44. [DOI] [PubMed] [Google Scholar]
- 214.Funakoshi A, Morikawa T, Muramatsu R, Yagi K, Seino M. A propsective WISC-R study in children with epilepsy. Jpn J Psychiatry Neurol. 1988. 1988;42(3):562–4. [DOI] [PubMed] [Google Scholar]
- 215.Rodin EA, Schmaltz S, Twitty G. Intellectual functions of patients with childhood-onset epilepsy. Developmental Medicine and Child Neurology. 1986. 1986;28:25–33. [DOI] [PubMed] [Google Scholar]
- 216.Berg AT, Lin J, Ebrahimi N, Testa FM, Levy SR, Shinnar S. Modeling remission and relapse in pediatric epilepsy: application of a Markov process. Epilepsy Res. 2004. June/2004;60(1):31–40. [DOI] [PubMed] [Google Scholar]
- 217.Mandelbaum DE, Burack GD, Bhise VV. Impact of antiepileptic drugs on cognition, behavior, and motor skills in children with new-onset, idiopathic epilepsy. Epilepsy Behav. 2009. October;16(2):341–4. [DOI] [PubMed] [Google Scholar]
- 218.Ijff DM, Aldenkamp AP. Cognitive side-effects of antiepileptic drugs in children. Handb Clin Neurol. 2013;111:707–18. [DOI] [PubMed] [Google Scholar]
- 219.Moavero R, Santarone ME, Galasso C, Curatolo P. Cognitive and behavioral effects of new antiepileptic drugs in pediatric epilepsy. Brain Dev. 2017. June;39(6):464–9. [DOI] [PubMed] [Google Scholar]
- 220.Trimble MR, Cull C. Children of school age: the influence of antiepileptic drugs on behavior and intellect. Epilepsia. 1988;29 Suppl 3:S15–9. [DOI] [PubMed] [Google Scholar]
- 221.Trimble MR, Cull CA. Antiepileptic drugs, cognitive function, and behavior in children. Cleve Clin J Med. 1989. 1989;56 Suppl Pt 1:S140–S6. [PubMed] [Google Scholar]
- 222.Farwell JR, Lee YJ, Hirtz DG, Sulzbacher SI, Ellenberg JH, Nelson KB. Phenobarbital for febrile seizures--effects on intelligence and on seizure recurrence. N Engl J Med. 1990. February/8/1990;322(6):364–9. [DOI] [PubMed] [Google Scholar]
- 223.Sulzbacher S, Farwell JR, Temkin N, Lu AS, Hirtz DG. Late cognitive effects of early treatment with phenobarbital. Clin Pediatr (Phila). 1999. July/1999;38(7):387–94. [DOI] [PubMed] [Google Scholar]
- 224.Kang HC, Eun BL, Wu Lee C, Ku Moon H, Kim JS, Wook Kim D, et al. The effects on cognitive function and behavioral problems of topiramate compared to carbamazepine as monotherapy for children with benign rolandic epilepsy. Epilepsia. 2007. September;48(9):1716–23. [DOI] [PubMed] [Google Scholar]
- 225.Coppola G, Verrotti A, Resicato G, Ferrarelli S, Auricchio G, Operto FF, et al. Topiramate in children and adolescents with epilepsy and mental retardation: a prospective study on behavior and cognitive effects. Epilepsy Behav. 2008. February;12(2):253–6. [DOI] [PubMed] [Google Scholar]
- 226.Brandl U, Kurlemann G, Neubauer B, Rettig K, Schäuble B, Schreiner A. Seizure and cognitive outcomes in children and adolescents with epilepsy treated with topiramate. Neuropediatrics. 2010. June;41(3):113–20. [DOI] [PubMed] [Google Scholar]
- 227.Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH, Van Emde BW, et al. Revised terminology and concepts for organization of seizures and epilepsies: report of the ILAE Commission on Classification and Terminology, 2005-2009. Epilepsia. 2010. April/2010;51(4):676–85. [DOI] [PubMed] [Google Scholar]
- 228.Freitag H, Tuxhorn I. Cognitive function in preschool children after epilepsy surgery: rationale for early intervention. Epilepsia. 2005. April/2005;46(4):561–7. [DOI] [PubMed] [Google Scholar]
- 229.Jonas R, Asarnow RF, LoPresti C, Yudovin S, Koh S, Wu JY, et al. Surgery for symptomatic infant-onset epileptic encephalopathy with and without infantile spasms. Neurology. 2005. February/22/2005;64(4):746–50. [DOI] [PubMed] [Google Scholar]
- 230.Lux AL, Edwards SW, Hancock E, Johnson AL, Kennedy CR, Newton RW, et al. The United Kingdom Infantile Spasms Study (UKISS) comparing hormone treatment with vigabatrin on developmental and epilepsy outcomes to age 14 months: a multicentre randomised trial. Lancet Neurol. 2005. November/2005;4(11):712–7. [DOI] [PubMed] [Google Scholar]
- 231.Beaumanoir A The Landau-Kleffner syndrome In: Roger J, Bureau M, Dravet C, Dreifuss FE, Perret A, Wolf P, editors. Epileptic Syndromes in Infancy, Childhood and Adolescence. 2 ed. London: John Libbey; 1992. p. 231–43. [Google Scholar]
- 232.Landau WM, Kleffner FR. Syndrome of acquired aphasia with convulsive disorder in children. Neurology. 1957. 1957;7:523–30. [DOI] [PubMed] [Google Scholar]
- 233.Hirsch E, Marescaux C, Maquet P, Metz-Lutz MN, Kiesmann M, Salmon E, et al. Landau-Kleffner Syndrome: a clinical and EEG study of five cases. Epilepsia. 1990. 1990;31:756–67. [DOI] [PubMed] [Google Scholar]
- 234.Lesca G, Rudolf G, Labalme A, Hirsch E, Arzimanoglou A, Genton P, et al. Epileptic encephalopathies of the Landau-Kleffner and continuous spike and waves during slow-wave sleep types: genomic dissection makes the link with autism. Epilepsia. 2012. September;53(9):1526–38. [DOI] [PubMed] [Google Scholar]
- 235.Tassinari CA, Rubboli G, Volpi L, Meletti S, d'Orsi G, Franca M, et al. Encephalopathy with electrical status epilepticus during slow sleep or ESES syndrome including the acquired aphasia. Clin Neurophysiol. 2000. September/2000;111 Suppl 2:S94–S102. [DOI] [PubMed] [Google Scholar]
- 236.Gencpinar P, Dundar NO, Tekgul H. Electrical status epilepticus in sleep (ESES)/continuous spikes and waves during slow sleep (CSWS) syndrome in children: An electroclinical evaluation according to the EEG patterns. Epilepsy Behav. 2016. August;61:107–11. [DOI] [PubMed] [Google Scholar]
- 237.Holmes GL. Interictal Spikes as an EEG Biomarker of Cognitive Impairment. Journal of Clinical Neurophysiology. 2020;in press. [DOI] [PubMed] [Google Scholar]
- 238.Sowell MK, Youssef PE. The Comorbidity of Migraine and Epilepsy in Children and Adolescents. Seminars in pediatric neurology. 2016. February;23(1):83–91. [DOI] [PubMed] [Google Scholar]
- 239.Rajapakse T, Buchhalter J. The borderland of migraine and epilepsy in children. Headache. 2016. June;56(6):1071–80. [DOI] [PubMed] [Google Scholar]
- 240.Mutlu A Association between epilepsy and headache. Neurol Sci. 2018. December;39(12):2129–34. [DOI] [PubMed] [Google Scholar]
- 241.Balottin L, Mannarini S, Candeloro D, Mita A, Chiappedi M, Balottin U. Rorschach Evaluation of Personality and Emotional Characteristics in Adolescents With Migraine Versus Epilepsy and Controls. Front Neurol. 2018;9:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Kanner AM, Ribot R, Mazarati A. Bidirectional relations among common psychiatric and neurologic comorbidities and epilepsy: Do they have an impact on the course of the seizure disorder? Epilepsia Open. 2018. December;3(Suppl Suppl 2):210–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Kossoff EH, Andermann F. Migraine and epilepsy. Seminars in pediatric neurology. 2010. June;17(2):117–22. [DOI] [PubMed] [Google Scholar]
- 244.Andermann F. Migraine and the benign partial epilepsies of childhood: evidence for an association. Epileptic Disord. 2000;2 Suppl 1:S37–9. [PubMed] [Google Scholar]
- 245.Papetti L, Nicita F, Parisi P, Spalice A, Villa MP, Kasteleijn-Nolst Trenité DG. "Headache and epilepsy"--how are they connected? Epilepsy Behav. 2013. March;26(3):386–93. [DOI] [PubMed] [Google Scholar]
- 246.Ludvigsson P, Hesdorffer D, Olafsson E, Kjartansson O, Hauser WA. Migraine with aura is a risk factor for unprovoked seizures in children. Ann Neurol. 2006. January;59(1):210–3. [DOI] [PubMed] [Google Scholar]
- 247.Ottman R, Lipton RB. Comorbidity of migraine and epilepsy. Neurology. 1994. 1994;44(Suppl. 7):5–10. [DOI] [PubMed] [Google Scholar]
- 248.Velioğlu SK, Boz C, Ozmenoğlu M. The impact of migraine on epilepsy: a prospective prognosis study. Cephalalgia. 2005. July;25(7):528–35. [DOI] [PubMed] [Google Scholar]
- 249.Kacperski J, Green A, Qaiser S. Management of Chronic Migraine in Children and Adolescents: A Brief Discussion on Preventive Therapies. Paediatr Drugs. 2020. September 5. [DOI] [PubMed] [Google Scholar]
- 250.Powers SW, Coffey CS, Chamberlin LA, Ecklund DJ, Klingner EA, Yankey JW, et al. Trial of Amitriptyline, Topiramate, and Placebo for Pediatric Migraine. N Engl J Med. 2017. January 12;376(2):115–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Loder E, Rizzoli P. Pharmacologic Prevention of Migraine: A Narrative Review of the State of the Art in 2018. Headache. 2018. November;58 Suppl 3:218–29. [DOI] [PubMed] [Google Scholar]
- 252.Kacperski J, Kabbouche MA, O'Brien HL, Weberding JL. The optimal management of headaches in children and adolescents. Ther Adv Neurol Disord. 2016. January;9(1):53–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Spritzer SD, Bravo TP, Drazkowski JF. Topiramate for Treatment in Patients With Migraine and Epilepsy. Headache. 2016. June;56(6):1081–5. [DOI] [PubMed] [Google Scholar]
- 254.Jackson JL, Cogbill E, Santana-Davila R, Eldredge C, Collier W, Gradall A, et al. A Comparative Effectiveness Meta-Analysis of Drugs for the Prophylaxis of Migraine Headache. PloS one. 2015;10(7):e0130733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Linde M, Mulleners WM, Chronicle EP, McCrory DC. Gabapentin or pregabalin for the prophylaxis of episodic migraine in adults. Cochrane Database Syst Rev. 2013. June 24;2013(6):Cd010609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Linde M, Mulleners WM, Chronicle EP, McCrory DC. Antiepileptics other than gabapentin, pregabalin, topiramate, and valproate for the prophylaxis of episodic migraine in adults. Cochrane Database Syst Rev. 2013. June 24(6):Cd010608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Patniyot IR, Gelfand AA. Acute Treatment Therapies for Pediatric Migraine: A Qualitative Systematic Review. Headache. 2016. January;56(1):49–70. [DOI] [PubMed] [Google Scholar]
- 258.Major PW, Grubisa HS, Thie NM. Triptans for treatment of acute pediatric migraine: a systematic literature review. Pediatric neurology. 2003. November;29(5):425–9. [DOI] [PubMed] [Google Scholar]
- 259.Jacob J, Goadsby PJ, Duncan JS. Use of sumatriptan in post-ictal migraine headache. Neurology. 1996. October;47(4):1104. [DOI] [PubMed] [Google Scholar]
- 260.Kanner AM. Management of psychiatric and neurological comorbidities in epilepsy. Nat Rev Neurol. 2016. February;12(2):106–16. [DOI] [PubMed] [Google Scholar]




