Neuro-Oncology
Anti-LGI1 encephalitis
Oct. 03, 2024
MedLink®, LLC
3525 Del Mar Heights Rd, Ste 304
San Diego, CA 92130-2122
Toll Free (U.S. + Canada): 800-452-2400
US Number: +1-619-640-4660
Support: service@medlink.com
Editor: editor@medlink.com
ISSN: 2831-9125
Toll Free (U.S. + Canada): 800-452-2400
US Number: +1-619-640-4660
Support: service@medlink.com
Editor: editor@medlink.com
ISSN: 2831-9125
Worddefinition
At vero eos et accusamus et iusto odio dignissimos ducimus qui blanditiis praesentium voluptatum deleniti atque corrupti quos dolores et quas.
Antiseizure medication (previously termed “antiepileptic drug”) treatment is the mainstay management of epilepsy. The laudable aim is freedom from seizures with minimal, if any, adverse medication reactions. This is achieved in about 50% to 70% of patients with a single, appropriately selected antiseizure medication at target therapeutic doses. This seizurefree rate varies significantly with seizure type and epileptic syndrome. Polytherapy should be avoided, if possible, but it is inevitable in about 30% to 50% of patients who fail to respond to single-medication therapy. Freedom of seizures should not be pursued at any cost and, in particular, at the expense of adverse medication reactions. The identification of adverse medication reactions, although sometimes difficult, is a crucial part of the management. The choice of antiseizure medication is demanding because an antiseizure medication beneficial for one type of epileptic seizure may be detrimental for another type of seizure. With the seemingly unstoppable development of a range of newer antiseizure medications over the past 2 decades, the available choice has been substantially widened, and the number of possible medication combinations for the treatment of epilepsy is almost limitless. Therefore, developing a framework to use antiseizure medications rationally has become an issue of important practical necessity. In this article, the author provides an updated overview of the pharmacotherapy of epilepsy in adolescents and adults, with particular reference to the clinical pharmacology of antiseizure medications, approaches to treatment, and principles of medication selection.
• Antiseizure medication treatment is generally recommended after two or more unprovoked seizures. | |
• The aim of antiseizure medication treatment is to achieve seizure freedom without adverse medication reactions. | |
• Selection of antiseizure medications should be individualized based on the seizure type or epilepsy syndrome, potential adverse effects, and medication-to-medication interactions. | |
• The first-option antiseizure medication should be the most likely to be efficacious and the most unlikely to cause adverse medication reactions. | |
• The correct antiseizure medication dose is the smallest one that achieves seizure control without adverse medication reactions; optimal effectiveness of an antiseizure medication may be lost by exceeding tolerability limits. | |
• An antiseizure medication appropriate for one type of seizure may be deleterious for another type. | |
• Two thirds of newly diagnosed patients will become seizure-free with antiseizure medication therapy, mostly when taking their first or second medication schedule, and often requiring no more than modest or moderate medication doses. | |
• The International League Against Epilepsy (ILAE) defines medication-resistant epilepsy as “failure of adequate trials of two tolerated and appropriately chosen and used antiseizure medication schedules (whether as monotherapies or in combination) to achieve sustained seizure freedom.” | |
• Patients with medication-resistant epilepsy should be referred to specialist centers for a comprehensive review of the diagnosis and management, and consideration of other therapeutic options, particularly epilepsy surgery. |
The history of the antiseizure medications is masterly detailed in a 2-part review by Shorvon published in Epilepsia (69; 70) and in a monograph giving a thorough account of how these medications were discovered and developed by Scott (67). See also a brief review by Brodie (13).
The contemporary antiseizure medication therapy started in 1857 with bromides, which were first mentioned in the English literature by Dr Edward Sieveking in his ‘‘Analyses of 52 cases of epilepsy observed by the author’’ (71). Although there are no randomized trials with bromide, there are enough clinical data to support its claim to be the first effective antiseizure medication. However, it is highly neurotoxic and became obsolete once better tolerated alternatives were found though bromide has support even to this day as a potential ‘‘medication of tertiary choice in the treatment of children with epilepsy’’ (13).
Modern pharmacotherapy of epilepsy was heralded by the serendipitous discovery of the anticonvulsant properties of phenobarbital in 1912 by Alfred Hauptmann (76). As a young resident psychiatrist, Hauptmann lived over a ward of people with epilepsy who were falling out of bed during the night because of tonic-clonic seizures, thereby, keeping him awake. Phenobarbital had been marketed the previous year as a hypnotic by F Bayer and Company. Hauptmann sedated his patients with phenobarbital so that he could get a good night’s sleep. Not only did his patients have fewer episodes during the night, but they did not have seizures on the following day. He published his observation, and the rest is history. Phenobarbital is still the most widely prescribed antiseizure medication in the developing world and remains a first popular choice in many industrialized countries, partly because of its modest cost (37; 76).
Phenytoin, the first nonsedating antiseizure medication, was introduced in the late 1930s as a result of systematic screening of compounds using novel animal seizure models. When Tracy Putnam was appointed to the directorship of the neurologic unit at Boston City Hospital in 1934, he set out to discover a less sedating antiseizure medication than phenobarbital. With the help of Frederic Gibbs, he established the first electroencephalographic laboratory for the routine study of ‘‘brain waves.” A makeshift apparatus was assembled to demonstrate that phenobarbital markedly raised the convulsive threshold in cats. Parke-Davis, and Company supplied Putnam’s research team with a number of non-sedating phenyl compounds. Only one of these, phenytoin was not too toxic for routine administration. Luckily, it was markedly effective in protecting cats from electrically induced convulsions. Putnam gave phenytoin to one of his young assistants, Houston Merritt, for clinical evaluation in 1936. The first patient to receive the medication had suffered daily seizures for many years and became permanently seizure-free on commencing treatment with phenytoin. The subsequent publication established this new medication in the therapeutic armamentarium. Phenytoin is still a widely used antiseizure medication in the United States.
During the 1940s, troxidone became established for the treatment of petit mal. Parke-Davis subsequently initiated a major research project to find a less toxic medication for this indication, which resulted in the licensing of ethosuximide in 1958. Its current value for absence seizures has been confirmed in a double-blind, randomized comparative trial with sodium valproate and lamotrigine (29).
The next major medication to be licensed was carbamazepine, which became widely available in the mid-1960s and is arguably supported by the best evidence base. It was synthesized by Schindler at Geigy in 1953 as a possible competitor for the antipsychotic chlorpromazine. The first study with carbamazepine in epilepsy was not carried out until 1963, after which it was rapidly licensed as an anticonvulsant in 1965 in the United Kingdom. That same year, the antiseizure activity of valproate was serendipitously recognized by Pierre Eymard while working as a research student at the University of Lyon. He dissolved a series of insoluble khellin and coumarin derivatives in valproate. Rather surprisingly, all of them appeared to have anticonvulsant properties. Subsequently valproate was subjected to extensive clinical investigation. Its sodium salt was first marketed as an antiseizure medication in France in 1967.
The value of the benzodiazepines for the treatment of epilepsy was rapidly recognized following their synthesis and development by Leo Sternbach while he was working for the Swiss pharmaceutical company, Roche, in the 1960s. In 1965, Henri Gastaut published a report regarding the efficacy of diazepam in treating status epilepticus. His follow-up paper with clonazepam 6 years later was even more positive. Clobazam is probably the most widely used oral benzodiazepine for a range of refractory epilepsies. Rectal and intravenous diazepam, buccal and intranasal midazolam, and lorazepam are medications of choice for acute repetitive seizures and convulsive status epilepticus.
The modern era of antiseizure medication development began in 1975 when the National Institute of Neurological Disorders and Stroke in the United States established the Anticonvulsant Drug Development Program. More than 28,000 chemical entities from academic and pharmaceutical chemists have since been screened, resulting in the licensing of an increasing list of antiseizure medications (70; 13). Thus, after a hiatus of nearly 20 years, there has been accelerated development of newer antiseizure medications, with the licensing of at least 16 compounds globally since the late 1980s. In chronological order, these were: vigabatrin, zonisamide, oxcarbazepine, lamotrigine, felbamate, gabapentin, topiramate, tiagabine, levetiracetam, pregabalin, rufinamide, stiripentol, lacosamide, eslicarbazepine, perampanel (70), and brivaracetam (26; 43). Twelve other antiseizure medications are in phase I-III clinical development (11), and still there are clear incentives to develop newer and more efficacious medications for epilepsy (23).
Brivaracetam | |
|
Treatment approach. The majority of newly diagnosed patients will have a good prognosis and become seizure-free with the first or second monotherapy, often requiring no more than modest or moderate medication doses (52; 06; 57; 14). They should be started on a single medication at low dose with increments over a number of weeks to establish an effective and tolerable regimen. Further dose adjustment can be made according to clinical response. An alternative medication is unavoidable when the patient develops intolerable adverse events. When and how to combine antiseizure medications remains controversial. Many authorities agree that it would be appropriate to consider combination therapy after two monotherapy attempts because the chance of success with a third medication is slim (17; 53; 07; 57; 31). Failure of two treatment schedules should prompt referral to a specialist center for further evaluation, including video-EEG telemetry to confirm the seizure or syndrome classifications, optimization of pharmacotherapy, and consideration of other therapeutic options, particularly epilepsy surgery.
Before starting prophylactic antiseizure medication in a patient with newly diagnosed seizures, a physician should be confident of the following:
1. The patient unequivocally has epileptic seizures. This requires definite exclusion of any imitators of epileptic seizures. | |
2. The epileptic seizures of the patient need treatment. This requires precise diagnosis of the epileptic syndrome and the type of seizures, their frequency and severity, their likelihood of relapse or remission, precipitating factors, and the patient and family concerns and understanding of the risks versus benefits of the antiseizure medication. Hard and fast rules are not always applicable. | |
3. The most appropriate antiseizure medication is selected for this particular patient with this particular type of seizure. The appropriate antiseizure medication is that which is the most likely of all others to be truly prophylactic as monotherapy (singlemedication treatment) for the seizures of the patient without causing undue adverse medication reactions. Carefully consider and exclude antiseizure medications that may worsen or be ineffective in this particular type of seizures. | |
4. The starting dose and titration of the selected medication should be in accordance with the appropriate recommendations, the age, and, primarily, the particular needs of the treated patient. |
All these should be thoroughly explained to the patient or guardian, and it should be ensured they fully understand.
Medication selection. Not only do antiseizure medications differ in the way they work, they vary substantially in their efficacy and side-effect profiles. The ideal profile of a firstchoice antiseizure medication in the prophylactic treatment of epilepsies is determined by a number of factors (Table 2). The importance of these factors varies significantly according to whether the antiseizure medication is used in monotherapy or polytherapy. A first-choice medication should have a spectrum of activity and side-effect and interaction profiles that has the potential to produce seizure freedom without intolerable toxicity and long-term sequelae for that individual. The choice should be matched to the patient's seizure type or epilepsy syndrome, age, gender, weight, psychiatric history, other disease states, concomitant medication, and lifestyle (16; 57). Therefore, this decision should be based on a thorough clinical history and physical examination aided by laboratory findings, including the EEG and brain imaging. The “prescribing information” (Food and Drug Administration in the United States) and the “summary of product characteristics” (European Medicines Agency in the European Union) are the most complete single sources of information on a medication. The summary of package characteristics can be obtained in any European language from http://www.ema.europa.eu/ema. In United Kingdom these are also available from http://emc. medicines.org.uk.
• Seizure specificity |
The antiseizure medication indications for the treatment of epileptic seizures according to the Food and Drug Administration package inserts are as follows. Please note that these indications are not the same with those of the European Medicines Agency and are often significantly different from the prevailing clinical practice (Table 3).
Brivaracetam. Treatment of partial onset seizures in patients 16 years of age and older with epilepsy.
Cannabidiol. Adjunctive treatment of seizures associated with two rare and severe forms of epilepsy: Lennox-Gastaut and Dravet syndrome, in patients aged 2 and older.
Carbamazepine
1. Partial seizures with complex symptomatology (psychomotor, temporal lobe). Patients with these seizures appear to show greater improvement than those with other types. | |
2. Generalized tonic-clonic seizures (grand mal). | |
3. Mixed seizure patterns that include the above or other partial or generalized seizures. Absence seizures (petit mal) do not appear to be controlled by carbamazepine |
Cenobamate. Treatment of partial-onset seizures in adults, either as monotherapy or as adjunctive therapy.
Clobazam. Adjunctive treatment of seizures associated with Lennox-Gastaut syndrome in patients 2 years of age or older.
Clonazepam. Adjunct in the treatment of the Lennox-Gastaut syndrome (petit mal variant) as well as akinetic and myoclonic seizures. In patients with absence seizures (petit mal) who have failed to respond to succinimides, clonazepam may be useful.
Divalproex sodium. Monotherapy and adjunctive therapy in the treatment of adult patients and pediatric patients as young as 10 years of age with:
1. Complex partial seizures that occur either in isolation or in association with other types of seizures. | |
2. Simple and complex absence seizures--and adjunctively in patients with multiple seizure types that include absence seizures. |
Eslicarbazepine acetate. Treatment of partial-onset seizures in patients 4 years of age and older.
Ethosuximide. Treatment of absence (petit mal) epilepsy.
Everolimus. Adjunctive treatment of adult and pediatric patients aged 2 and older with tuberous sclerosis complex–associated partial-onset seizures.
Felbamate. Felbamate is not indicated as a first-line antiseizure treatment. Felbamate is recommended for use only in those patients who respond inadequately to alternative treatments and whose epilepsy is so severe that a substantial risk of aplastic anemia or liver failure is deemed acceptable in light of the benefits conferred by its use.
If these criteria are met and the patient has been fully advised of the risk and has provided written acknowledgement, felbamate can be considered for either monotherapy or adjunctive therapy in the treatment of partial seizures, with and without generalization, in adults with epilepsy and as adjunctive therapy in the treatment of partial and generalized seizures associated with Lennox-Gastaut syndrome in children.
Fenfluramine. Treatment of seizures associated with Dravet syndrome in patients aged 2 and older.
Gabapentin. Adjunctive therapy in the treatment of:
1. Partial seizures with and without secondary generalization in patients over 12 years of age with epilepsy. | |
2. Partial seizures in pediatric patients aged 3 to 12 years. |
Ganaxolone. Treatment of seizures associated with cyclin-dependent kinase-like 5 deficiency disorder in patients 2 years of age and older.
Lacosamide. Monotherapy or adjunctive therapy in patients 17 years and older with partial-onset seizures as adjunctive therapy. Injection for intravenous use is an alternative when oral administration is temporarily not feasible.
Lamotrigine. Adjunctive therapy for the following seizure types in patients 2 years of age or older: partial seizures, primary generalized tonic-clonic seizures, generalized seizures of Lennox-Gastaut syndrome.
Conversion to monotherapy in adults (16 years of age and older) with partial seizures who are receiving treatment with carbamazepine, phenytoin, phenobarbital, primidone, or valproate as the single antiseizure medication.
Safety and effectiveness of lamotrigine tablets have not been established (1) as initial monotherapy; (2) for conversion to monotherapy from antiseizure medications other than carbamazepine, phenytoin, phenobarbital, primidone, or valproate; or (3) for simultaneous conversion to monotherapy from two or more concomitant antiseizure medications.
Levetiracetam. Adjunctive therapy in the treatment of:
1. Partial onset seizures in adults and children 4 years of age and older with epilepsy. | |
2. Myoclonic seizures in adults and adolescents 12 years of age and older with juvenile myoclonic epilepsy. | |
3. Primary generalized tonic-clonic seizures in adults and children 6 years of age and older with idiopathic generalized epilepsy. |
Oxcarbazepine.
1. Monotherapy or adjunctive therapy in the treatment of partial seizures in adults. | |
2. Monotherapy in the treatment of partial seizures in children aged 4 years and above. | |
3. Adjunctive therapy in children aged 2 years and above with partial seizures. |
Perampanel.
1. Treatment of partial-onset seizures with or without secondarily generalized seizures in patients with epilepsy 12 years of age and older. | |
2. Adjunctive therapy for the treatment of primarily generalized tonic-clonic seizures in patients with epilepsy 12 years of age and older. |
Phenobarbital. Treatment of generalized and partial seizures.
Phenytoin. Treatment of tonic-clonic (grand mal) and psychomotor (temporal lobe) seizures and prevention and treatment of seizures occurring during or following neurosurgery.
Pregabalin. Adjunctive therapy for adult patients with partial onset seizures.
Primidone. Monotherapy or adjunctive treatment of grand mal, psychomotor, and focal epileptic seizures. It may control grand mal seizures refractory to other anticonvulsant therapy.
Rufinamide. Adjunctive treatment of seizures associated with Lennox-Gastaut syndrome in pediatric patients 1 year of age and older and in adults.
Stiripentol. Adjunctive treatment of seizures associated with Dravet syndrome in patients aged 2 and older who are taking clobazam.
Tiagabine hydrochloride. Adjunctive therapy in adults and children 12 years and older in the treatment of partial seizures.
Topiramate.
1. Initial monotherapy in patients 2 years of age and older with partial onset or primary generalized tonic-clonic seizures. | |
2. Adjunctive therapy for adults and pediatric patients ages 2 to 16 years with partial onset seizures or primary generalized tonic-clonic seizures, and in patients 2 years of age and older with seizures associated with Lennox-Gastaut syndrome. |
Valproic acid.
1. Monotherapy and adjunctive therapy in the treatment of patients with complex partial seizures that occur either in isolation or in association with other types of seizures. | |
2. Monotherapy and adjunctive therapy in the treatment of simple and complex absence seizures. | |
3. Adjunctively in patients with multiple seizure types, including absence seizures. |
Vigabatrin.
1. Adjunctive therapy for adults and pediatric patients 10 years of age and older with refractory complex partial seizures who have inadequately responded to several alternative treatments and for whom the potential benefits outweigh the risk of vision loss. It is not indicated as a first-line agent for complex partial seizures. | |
2. Monotherapy for pediatric patients with infantile spasms 1 month to 2 years of age for whom the potential benefits outweigh the potential risk of vision loss. |
Zonisamide. Adjunctive therapy in the treatment of partial seizures in adults with epilepsy.
Antiseizure medications |
Focal seizures (simple or complex) |
Secondarily GTCSs |
Primarily GTCSs |
Myoclonic jerks |
Absence seizures |
Brivaracetam |
Effective |
Effective |
Unknown |
Effective? |
Unknown |
Cannabidiol |
Effective in tuberous sclerosis complex |
Effective in tuberous sclerosis complex |
Effective in tuberous sclerosis complex, Dravet syndrome, Lennox-Gastaut syndrome |
Unknown |
Unknown |
Carbamazepine |
Effective |
Effective |
Effective |
May exacerbate |
May exacerbate |
Cenobamate |
Effective |
Effective |
Unknown |
Unknown |
Unknown |
Clobazam |
Effective |
Effective |
Effective? |
Effective? |
Effective? |
Clonazepam |
Effective? |
Effective? |
Ineffective? |
Effective |
Effective |
Eslicarbazepine |
Effective |
Effective |
Unknown |
Unknown |
Unknown |
Ethosuximide |
- |
- |
- |
Effective? |
Effective |
Everolimus |
Effective in tuberous sclerosis complex |
- |
Unknown |
Unknown |
Unknown |
Fenfluramine |
Unknown |
Unknown |
Effective in Dravet syndrome and Lennox-Gastaut syndrome, especially drop attacks |
Unknown |
Unknown |
Gabapentin |
Effective |
Effective |
- |
- |
May exaggerate |
Ganaxolone |
Unknown |
Unknown |
Effective for epileptic spasms in CDKL5 deficiency |
Unknown |
Unknown |
Lacosamide |
Effective |
Effective |
Unknown |
May exacerbate |
Unknown |
Lamotrigine |
Effective |
Effective |
Effective |
May exacerbate |
Effective |
Levetiracetam |
Effective |
Effective |
Effective |
Effective |
Effective |
Oxcarbazepine |
Effective |
Effective |
Effective |
- |
- |
Perampanel |
Effective |
Effective |
Effective |
Unknown |
Unknown |
Phenobarbital |
Effective |
Effective |
Effective |
Effective |
Effective |
Phenytoin |
Effective |
Effective |
Effective |
Effective |
- |
Pregabalin |
Effective |
Effective |
- |
Exacerbates |
- |
Stiripentol |
Unknown |
Unknown |
Effective in Dravet |
Unknown |
Unknown |
Tiagabine |
Effective |
Effective |
- |
- |
Exacerbates |
Topiramate |
Effective |
Effective |
Effective |
Effective? |
Effective? |
Valproate |
Effective |
Effective |
Effective |
Effective |
Effective |
Vigabatrin |
Effective |
Effective |
- |
- |
May exacerbate |
Zonisamide |
Effective |
Effective |
Effective? |
Effective? |
Effective? |
|
Medication |
Starting dose (mg/day) |
Commonest dose (mg/day) |
Maintenance dose (mg/day) |
Usual dosing interval |
Established antiseizure medications | ||||
Carbamazepine |
200 |
600 |
800-1200 |
2 times daily |
Newer antiseizure medications | ||||
Brivaracetam |
50 |
100 |
100-200 |
2 times daily |
Recently approved antiseizure medications | ||||
Cannabidiol |
5 mg/kg |
10 mg/kg |
10-20 mg/kg |
2 times daily |
Cenobamate |
12.5 |
200 |
200-400 |
1 time daily |
Everolimus |
5 mg/m2 |
Depends on trough |
Depends on trough |
1 time daily |
Fenfluramine |
0.2 mg/kg |
0.7 mg/kg |
0.2-0.7 mg/kg |
2 times daily |
Ganaxolone |
6 mg/kg or 150 mg if >28 kg |
21 mg/kg or 600 mg if >28 kg |
21 mg/kg or 600 mg if >28 kg |
3 times daily |
Stiripentol |
5 mg/kg |
20-30 mg/kg |
20-50 mg/kg |
2-3 times daily |
|
Efficacy and tolerability. The profile of activity against different seizure types varies among the antiseizure medications (Table 3). In addition, certain epilepsy syndromes have been found to be particularly responsive to specific antiseizure medications. For instance, juvenile myoclonic epilepsy responds well to valproate and levetiracetam, whereas vigabatrin is regarded by many as the medication of choice for infantile spasms. A double-blind randomized controlled trial showed that ethosuximide and valproic acid were more effective than lamotrigine for childhood absence epilepsy (29). Therefore, it is of paramount importance to accurately classify the seizure type and epilepsy syndrome and choose the most appropriate medication for that individual.
Systematic reviews of randomized, controlled trials comparing phenobarbital, primidone, phenytoin, carbamazepine, and valproate for partial and tonic-clonic seizures identified no significant difference in efficacy among these established antiseizure medications, with the possible exception of a small benefit of carbamazepine over valproate for partial seizures (46). Phenobarbital and primidone produced higher withdrawal rates due to their sedative effect, although these medications are often well tolerated at moderate dosing in clinical practice.
None of the newer antiseizure medications have demonstrated superior efficacy when tested against the established agents in monotherapy studies for the treatment of newly diagnosed partial and tonic-clonic seizures. Evidence of better tolerability has been shown in some cases, in particular fewer neurotoxic side effects. Thus, lamotrigine and oxcarbazepine showed better overall effectiveness than carbamazepine and phenytoin, respectively.
Based on the findings from randomized, controlled trials, various national and international guidelines have been developed for the use of antiseizure medications as initial monotherapy for epilepsy. These guidelines focus on the efficacy and effectiveness aspects in the process of medication selection (25; 28; 55).
These guidelines are valuable in providing a working framework for the treatment of new-onset epilepsy. The differences in their recommendations may reflect fundamental differences in the purposes of the guidelines and in the approaches adopted by the groups developing them. It is important to bear in mind that these treatment guidelines focus on short-term efficacy and effectiveness evidence alone, and other patient-related factors, discussed below, must be considered when choosing the most appropriate treatment for the individual patient. Continuous advances in medication development also mean that these guidelines become rapidly outdated.
Medication |
Absorption (bioavailability %) |
Protein binding |
Elimination half-life (hours) |
Routes of elimination |
Older antiseizure medications | ||||
Carbamazepine |
Slow (75-80) |
70-80 |
24-45 (single) |
Hepatic metabolism |
Clobazam |
Rapid (90-100) |
87-90 |
10-30 |
Hepatic metabolism |
Clonazepam |
Rapid (80-90) |
80-90 |
30-40 |
Hepatic metabolism |
Ethosuximide |
Rapid (90-95) |
0 |
20-60 |
Hepatic metabolism |
Phenobarbital |
Slow (95-100) |
48-54 |
72-144 |
Hepatic metabolism |
Phenytoin |
Slow (85-90) |
90-93 |
9-40 |
Saturable hepatic metabolism |
Primidone |
Rapid (90-100) |
20-30 |
4-12 |
Hepatic metabolism |
Valproate |
Rapid (100) |
88-92 |
7-17 |
Hepatic metabolism |
Newer antiseizure medications | ||||
Brivaracetam |
Rapid (100) |
≤ 20% |
9 |
Hydrolysis |
Eslicarbazepine acetate ‡ |
Rapid (90) |
40 |
13 - 20 |
Glucuronidation |
Felbamate |
Slow (95-100) |
22-36 |
13-23 |
Hepatic metabolism |
Gabapentin ‡ |
Slow (60) |
0 |
6-9 |
Not metabolized |
Lacosamide ‡ |
Rapid (95-100) |
< 15 |
13 |
Hepatic metabolism |
Lamotrigine |
Rapid (95-100) |
55 |
22-36 |
Glucuronidation |
Levetiracetam |
Rapid (95-100) |
< 10 |
7-8 |
Non-hepatic hydrolysis |
Oxcarbazepine |
Rapid (95-100) |
40† |
8-10† |
Hepatic conversion to active moiety |
Perampanel |
Rapid (95-100) |
95 |
105 |
Hepatic primary oxidation and sequential glucuronidation |
Pregabalin ‡ |
Rapid (90-100) |
0 |
6 |
Renal excretion |
Rufinamide |
Slow |
34 |
6-10 |
Hepatic metabolism |
Tiagabine ‡ |
Rapid (95-100) |
96 |
5-9 |
Hepatic metabolism |
Topiramate |
Slow (80) |
9-17 |
20-24 |
Hepatic metabolism |
Vigabatrin |
Slow (60-80) |
0 |
5-7 |
Not metabolized |
Zonisamide |
Rapid (95-100) |
40-60 |
50-68 |
Hepatic metabolism |
Recently approved antiseizure medications | ||||
Cannabidiol |
Slow (non-linear dose related) |
>94 |
56-61 |
Hepatic metabolism |
Cenobamate |
Fast (> 88%) |
60 |
50-60 |
Hepatic metabolism |
Everolimus |
Slow (5-10) |
75 |
Varies by tissue |
Hepatic metabolism |
Fenfluramine |
Slow (70-85) |
50 |
24-30 |
Hepatic metabolism |
Ganaxolone |
Rapid (10) |
99 |
34 |
Hepatic metabolism |
Stiripentol |
Rapid (10-30) |
99 |
5-13 |
Hepatic metabolism |
Polytherapy. Given that most patients respond to their first or second antiseizure medication, combination therapy should usually be considered after failure of two monotherapy regimens due to lack of efficacy. If poor tolerability is the problem, a third monotherapy could reasonably be tried. Some patients experiencing many seizures immediately before diagnosis may be best treated with combination therapy following a partial response to the first antiseizure medication, particularly if an underlying anatomical problem, such as cortical dysplasia or hippocampal atrophy, has been identified (38).
When making a decision about polytherapy, one should first scrutinize the possible or probable reasons why the monotherapy failed. The following possibilities, which often require reevaluation of diagnosis (genuine epileptic seizures? what type of seizures?), would be considered (57):
• The patient does not have epileptic seizures. | |
• The patient has both epileptic and nonepileptic seizures. | |
• The patient has focal and not generalized seizures or vice versa. | |
• The antiseizure medication used as monotherapy was not suitable for the particular type of seizures in this patient because of contraindications (tiagabine or carbamazepine in absences or myoclonic jerks and lamotrigine in myoclonic epilepsies), weak efficacy (valproate or GABA in in focal seizures), or total ineffectiveness (gabapentin in primarily GTCSs). | |
• The patient is not adherent to treatment. Non-adherence varies from unwillingness to take medication to occasionally forgetting or missing the antiseizure medication dose or violating particularly eminent seizureprecipitating factors, such as photic stimulation, sleep deprivation, and alcohol or medication abuse. |
The ideal profile of an antiseizure medication for polypharmacy includes all the factors that are important for monotherapy (Table 2), but with particular emphasis on the following points (57):
Strength of efficacy. This may be increased or weakened by pharmacokinetic and pharmacodynamic interactions.
Safety and tolerability. These are often worsened by pharmacokinetic and pharmacodynamic interactions.
Medication interactions. Interactions with other antiseizure medications, whether pharmacokinetic, pharmacodynamic, or both, are particularly unwanted in polytherapy. Raising the levels of concomitant antiseizure medications and pharmacodynamic interactions may lead to toxic effects. Conversely, decreasing their levels may increase and worsen seizures causing a vicious cycle in clinical management. With the exception of lacosamide, levetiracetam, and gabapentin, most of the newer antiseizure medications exhibit sometimes complex, undesirable, medication-to-medication interactions. For example, consider that lamotrigine:
• requires different dosage and titration schemes when combined with hepatic enzyme inducers and when combined with valproate. | |
• pharmacodynamic interactions enhance toxicity and teratogenicity (although pharmacodynamic interactions have a beneficial effect on efficacy). | |
• levels lower more than half during pregnancy or hormonal contraception. |
Different mechanisms of action in relation to other concurrent antiseizure medications (Table 8). Antiseizure medication-to-medication interactions may be purely additive, antagonistic, or synergistic. Antiseizure medications with the same mechanism of action would be expected to be additive, whereas combining antiseizure medications with different mechanisms of action may have synergistic effects. A sodium channel blocker antiseizure medication combined with another that increases the GABAergic neurotransmission or that has multiple mechanisms is generally more effective than a combination of two sodium channel blockers (21; 17; 39). An antiseizure medication is unlikely to have better success and more likely to have additive adverse medication reactions if added to another antiseizure medication with the same mechanism of action.
The risks of polytherapy include:
• more adverse medication reactions |
Converting from polytherapy to monotherapy. Evidence from studies with older and newer antiseizure medications shows that a significant number of patients can be converted successfully from polytherapy to monotherapy without losing seizure control and, in some cases, with improved seizure control. In these cases, the antiseizure medication that appears, after careful consideration, to be the least effective is gradually withdrawn. “Gradually” sometimes means in steps of weeks or months. This should be particularly slow for certain antiseizure medications, such as phenobarbital and benzodiazepines, in order to avoid withdrawal seizures.
Medication-to-medication interactions. Antiseizure medication interactions are an important consideration in the treatment of epilepsy and, indeed, can be a major therapeutic challenge. The principal pharmacokinetic interaction relates to hepatic enzyme induction or inhibition whilst pharmacodynamic interactions principally entail adverse effect synergism, although examples of anticonvulsant synergism also exist. The older antiseizure medications are commonly associated with pharmacokinetic interactions by inducing the synthesis of a range of monooxygenase and conjugating enzymes (phenobarbital, primidone, phenytoin, and carbamazepine, in particular, accelerate the breakdown of many commonly prescribed lipid-soluble medications, including oral contraceptives and warfarin). Valproate is a weak enzyme inhibitor, which can slow the clearance of other antiseizure medications, such as phenytoin and lamotrigine. Hepatic enzyme induction stimulates the production and increases the amount of CYP enzymes. This, in turn, increases the rate of metabolism of medications metabolized by the CYP enzymes, thus, resulting in lower plasma concentrations. Conversely, hepatic enzyme inducers may increase the bioavailability of medications that require metabolism for their activation. The effect of hepatic induction, unlike hepatic inhibition persists for several days following the withdrawal of the enzyme-inducing medication.
Some of the new antiseizure medications are less interacting primarily because many are renally excreted or not hepatically metabolized (eg, gabapentin, lacosamide, levetiracetam, topiramate, vigabatrin) and most do not (or minimally) induce or inhibit hepatic metabolism. The least pharmacokinetic interactions are associated with gabapentin, lacosamide, tiagabine, vigabatrin, and zonisamide, whereas lamotrigine, felbamate, oxcarbazepine, and rufinamide are associated with the most. To date, felbamate, gabapentin, oxcarbazepine, perampanel, pregabalin, rufinamide, stiripentol, and zonisamide have not been associated with any pharmacodynamic interactions (58).
Pharmacodynamics refers to the biochemical and physiological effects of medications and their mechanisms of action. They may be additive, synergistic, or antagonistic. They may be beneficial, detrimental or both, as exemplified by lamotrigine combined with valproate (increased therapeutic efficacy but also increased risk of adverse medication reactions and teratogenicity).
Therapeutic medication monitoring. Monitoring of circulating concentrations of antiseizure medications became possible in the late 1960s, initially as a research tool but was rapidly adopted for widespread clinical use largely because phenytoin was shown to undergo saturation kinetics (63). It is important to bear in mind that the commonly quoted “therapeutic” or “reference” range has a probabilistic meaning; that is, it refers to the serum concentrations at which the majority of patients can be expected to show a response without undue adverse effects. Some patients will do well with medication concentrations below the lower limit of the range whereas others will tolerate amounts exceeding the upper level with benefit and without toxicity (59; 42). Therefore, the dose should not be increased or reduced just to attain a defined “therapeutic” concentration in patients who are seizure- and side-effect-free.
The ILAE position paper of 2008 is a practice guideline for therapeutic medication monitoring and is highly recommended for further reading (59). It also provides pharmacokinetic details, interactions with other antiseizure medications, and reference ranges for each antiseizure medication and discusses the role of therapeutic medication monitoring in children and the elderly and during pregnancy.
Serum medication monitoring is best performed with a specific clinical question in mind. Situations in which serum medication monitoring are most likely to be of benefit include: (1) when a person has attained the desired clinical outcome, to establish an individual therapeutic concentration that can be used at subsequent times to assess potential causes for a change in medication response; (2) as an aid in the diagnosis of clinical toxicity; (3) to assess compliance; (4) to guide dosage adjustment in situations associated with increased pharmacokinetic variability (eg, children, the elderly, patients with associated diseases, medication formulation changes); (5) when a potentially important pharmacokinetic change is anticipated (eg, in pregnancy or when an interacting medication is added or removed); (6) to guide dose adjustments for antiseizure medications with dose-dependent pharmacokinetics, particularly phenytoin (59).
Monitoring the circulating concentration is of greatest value with agents that exhibit nonlinear pharmacokinetics, in particular phenytoin. Increases and decreases in dosing may result in disproportionately large changes in serum concentration. Substantial variation in carbamazepine levels over the course of a day--as much as 100% with twice-daily dosing--makes interpretation problematic unless the timing of dosing and blood sampling are standardized. Because valproate does not exhibit a clear-cut concentration-effect-toxicity relationship, monitoring of this medication is not very helpful. Concentration-response-toxicity studies with the newer antiseizure medications have not demonstrated consistent correlations between circulating levels and efficacy or side effects (32).
Trough antiseizure medication plasma levels are important with regard to efficacy, whereas peak antiseizure medication plasma levels are important with regard to toxicity.
Prescription of brand-to-generic or generic-to-generic antiseizure medication. There is considerable concern among physicians and patients about the efficacy and safety of brand-to-generic antiseizure medication substitution or vice versa and also from one to another generic medication of different manufacturers (50). These concerns have been reasonably raised because of well-documented breakthrough seizures, emergency-treated epilepsy events, and toxicity as a result of such substitutions.
Substitution from one to another antiseizure medication of the same active substance is safe only when these are of therapeutic equivalence; medication products are considered to be therapeutic equivalents if they can be expected to have the same clinical effect and safety profile when administered to patients under the conditions specified in the labelling. However, even when the generic product is from a reliable source (something that is beyond the role of prescribing physicians) there may be problems with their therapeutic equivalence, particularly in terms of bioavailability and pharmacokinetic profiles.
Antiseizure medications are particularly vulnerable in these changes because many have a narrow reference concentration index between medication levels that are therapeutic and those that may cause adverse medication reactions.
“For antiepileptic drugs, small variations in concentrations between name brands and their generic equivalents can cause toxic effects or seizures when taken by patients with epilepsy. The American Academy of Neurology believes that the authorities should give complete physician autonomy in prescribing antiepileptic drugs” (50). It is because of these problems that formal recommendations worldwide uniformly warn against such substitutions of antiseizure medications.
A patient who is stabilized with an antiseizure medication in terms of seizure control and minimal adverse medication reactions should remain in the same brand or the particular generic product; interchanging products introduces risk.
When an antiseizure medication is first prescribed, it can be a brand name or a generic product providing that the latter has been truly tested in regard to the credibility of its manufacturers and bioequivalence with the brand product. Titration and maintenance should be with the same antiseizure medication product.
It is unreasonable to switch from one to another antiseizure medication, either from an expensive branded antiseizure medication to a cheaper bioequivalent generic product or vice versa. Switching is unnecessary and may impose significant risk to the patient. When such substitution is attempted, the patient should be well informed of possible consequences, such as seizure relapse or adverse medication reactions.
Prescriptions should clearly indicate the type of antiseizure medication formulation to be used, even for generic products (20; 57; 61). However, generic antiseizure medications with Food and Drug Administration–validated bioequivalence appears to be a safe clinical choice (33; 62).
Depending on study methodology and inclusion criteria, the chance of recurrence after a single unprovoked seizure reported in the literature ranges from 23% to 71% (09). Because a substantial proportion of patients will not have further episodes, and randomized studies suggest that early treatment does not improve prognosis (54; 45), most specialists do not recommend antiseizure medication therapy after a single seizure. However, when the chance of recurrence is high, eg, in the presence of an underlying cerebral lesion or epileptiform discharge on EEG, treatment may commence after the first tonic-clonic seizure.
Patients reporting more than one well-documented or witnessed seizure usually require treatment. Exceptions can include widely separated events, provoked seizures (eg, concomitant illness, photosensitive epilepsy, alcohol withdrawal), and benign childhood epilepsy syndromes. In addition, there is little chance of successful treatment in patients such as alcohol abusers, drug addicts, and conscientious objectors who are unlikely or unwilling to take medication. The decision of whether or not to start treatment should be made after ample discussion with the patient and his or her family of the risks and benefits of both courses of action.
Patients whose seizures are nonepileptic, including psychogenic/functional or “pseudoseizures,” should not be treated with antiseizure medications. Those whose seizures can be prevented by avoidance of provoking factors such as alcohol, illicit drugs, sleep deprivation, or flashing lights may not require long-term antiseizure medication therapy.
An antiseizure medication is contraindicated not only when it exaggerates seizures but also when it is ineffective in controlling the seizures it is supposed to treat. It may cause unnecessary adverse medication reactions and deprive the patient of the therapeutic effect that could be provided by another appropriate antiseizure medication (57).
Treatment with an antiseizure medication may fail due to lack of efficacy, intolerable adverse effects, or both. In the Glasgow series of 780 newly diagnosed patients, 47% became seizure-free with the first monotherapy, over 80% of whom required no more than modest to moderate doses. Another 10% responded to the second monotherapy. Only 2.3% of the cohort entered remission with the third monotherapy or with polytherapy (52; 53). Results from this and other studies suggest that failure of the first medication due to lack of efficacy is a powerful predictor of subsequent refractoriness and that the probability of remission after the failure of two to three successive medication regimens is slim (35; 53).
When the newer antiseizure medications were studied as add-on treatment in randomized, controlled trials in refractory epilepsy, a reduction in seizure frequency by 50% from the baseline was achieved in only 30% to 50% of patients. The clinical relevance of a 50% reduction in seizure frequency has been called into question (51). Studies including patients treated surgically (27) or medically (12) suggest that absolute seizure freedom is the only outcome consistently associated with improvement in quality of life. A meta-analysis showed that the overall weighted pooled-risk difference in favor of the newer antiseizure medications over placebo for seizure-freedom during the limited study periods was only 6% (10).
Adverse effects are a leading cause of treatment failure with antiseizure medications. They result in early treatment discontinuation in up to 25% of patients and may preclude attainment of fully effective doses and have a negative effect on patient adherence. Furthermore, adverse effects of antiseizure medications are a major source of disability, morbidity, and mortality and a substantial burden on use and costs of health care. Substantial effort has been made to define, quantify, and address their clinical relevance (65; 60).
According to the World Health Organization classification, there are five types of medication-related adverse effects:
1. Acute, related to the pharmacological properties of the medication (type A) |
Some of the more common or serious adverse effects are shown in Table 6. Serious and life-threatening adverse effects are highlighted by the Food and Drug Administration in boxed warnings of the package inserts.
Antiseizure medication |
Primary adverse reactions |
Life threatening? |
Brivaracetam |
Somnolence, dizziness, fatigue |
New medication with few exposures |
Carbamazepine |
Idiosyncratic (rash), sedation, headache, ataxia, nystagmus, diplopia, tremor, impotence, hyponatremia, cardiac arrhythmia |
Antiseizure medication hypersensitivity syndrome (AHS), hepatic failure, hematological |
Clobazam |
Severe sedation, fatigue, drowsiness, behavioral and cognitive impairment, restlessness, aggressiveness, hypersalivation, coordination disturbances, tolerance, withdrawal syndrome |
No |
Clonazepam |
As for clobazam |
No |
Eslicarbazepine |
Idiosyncratic (rash), dizziness, somnolence, headache, ataxia, inattention, diplopia, tremor, nausea, vomiting, hyponatremia, PR prolongation in ECG |
No |
Ethosuximide |
Idiosyncratic (rash), gastrointestinal disturbances, anorexia, weight loss, drowsiness, photophobia, headache |
AHS, renal and hepatic failure, hematological |
Gabapentin |
Weight gain, peripheral edema, behavioral changes, impotence, viral infection |
Acute pancreatitis, hepatitis, Stevens-Johnson syndrome, acute renal failure‡ |
Lacosamide |
Dizziness, headache, diplopia, nausea, vomiting, blurred vision, PR prolongation in ECG |
No |
Lamotrigine |
Idiosyncratic (rash), tics, insomnia, dizziness, diplopia, headache, ataxia, asthenia |
AHS, hepatic failure, hematological |
Levetiracetam |
Irritability, behavioral and psychotic changes, asthenia, dizziness, somnolence, headache |
Hepatic failure, hepatitis |
Oxcarbazepine |
Idiosyncratic (rash), headache, dizziness, weakness, nausea, somnolence, ataxia and diplopia, hyponatremia |
AHS, hematological |
Perampanel |
Serious or life-threatening psychiatric and behavioral adverse reactions including aggression, hostility, irritability, anger, and homicidal ideation and threats. Dizziness, gait disturbances, fatigue, falls, weight gain |
Yes |
Phenobarbital |
Idiosyncratic (rash), severe drowsiness, sedation, impairment of cognition and concentration, hyperkinesia and agitation in children, shoulder-hand syndrome |
AHS, hepatic failure, hematological |
Phenytoin |
Idiosyncratic (rash), ataxia, drowsiness, lethargy, sedation, encephalopathy, gingival hyperplasia, hirsutism, dysmorphism, rickets, osteomalacia |
AHS, hepatic failure, hematological |
Pregabalin |
Weight gain, myoclonus, dizziness, somnolence, ataxia, confusion |
Renal failure, congestive heart failure, AHS |
Tiagabine |
Stupor or spike-wave stupor, weakness |
Status epilepticus |
Topiramate |
Somnolence, anorexia, fatigue, nervousness, difficulty with concentration or attention, memory impairment, psychomotor slowing, metabolic acidosis, weight loss, language dysfunction, renal calculi, acute angle-closure glaucoma and other ocular abnormalities, paresthesia |
Hepatic failure, anhidrosis |
Valproate |
Nausea, vomiting, dyspepsia, weight gain, tremor, hair loss, hormonal issues in women |
Hepatic and pancreatic failure |
Vigabatrin |
Irreversible visual field defects, fatigue, weight gain |
No |
Zonisamide |
Idiosyncratic, drowsiness, anorexia, irritability, photosensitivity, weight loss, renal calculi |
AHS, anhidrosis |
Newer antiseizure medications |
Primary adverse reactions |
Life threatening? |
Cannabidiol |
Somnolence, fatigue, weight loss, thrombocytopenia |
Status epilepticus, hepatotoxicity |
Cenobamate |
Sleepy, dizziness, double vision, headache |
DRESS (medication reaction with eosinophilia and systemic symptoms), QT shortening |
Everolimus |
Delayed wound healing, tiredness, stomach pain, nausea, headache, decreased appetite |
Breathing problems, infection, renal failure |
Fenfluramine |
Different side effect profiles in Lennox-Gastaut syndrome vs. Dravet syndrome; both have diarrhea, sleepiness, vomiting. Dravet syndrome: decreased weight, drooling |
Status epilepticus, valvular heart disease |
Ganaxolone |
Somnolence, sedation, upper respiratory tract infection, fever, loss of voice, vision loss, tachycardia |
None |
Stiripentol |
Aggression, headache, fever, trouble breathing, insomnia, worse motor control |
None |
The table above is based predominantly on information obtained from the summaries of product characteristics (SmPCs3-19) and package inserts (57). It is an assessment of common adverse medication reactions and those of clinical importance. It is not an exhaustive list of all adverse medication reactions; for these, readers should refer to the summaries of product characteristics or package inserts of each antiseizure medication. There is a comprehensive review of the safety profile of the newest antiseizure medications: brivaracetam, eslicarbazepine acetate lacosamide, and perampanel (56).
Type A effects can be ascribed to the known mechanism of action of the medication; they usually arise at the beginning of treatment or after dose escalation and typically abate over time or after dose reduction. The most representative type A effects of antiseizure medications affect the CNS and include drowsiness, fatigue, dizziness, unsteadiness, blurred or double vision, difficulty concentrating, memory problems, irritability, and depression. These are often dose-related and can be anticipated by adopting a slow-titration schedule. It is unclear why some patients develop intolerable neurotoxic side effects at starting doses whereas others can tolerate substantial amounts of the same agent with impunity (16). Genetic factors likely play a role.
Type B effects (idiosyncratic). The most common idiosyncratic reaction to antiseizure medications is skin rash, which can range from a trivial evanescent exanthem to life-threatening severe cutaneous reactions (Stevens-Johnson syndrome and toxic epidermal necrolysis) (04). Maculopapular rashes are noted in 5% to 17% of patients started on carbamazepine, phenytoin, phenobarbital, and lamotrigine. Severe mucocutaneous reactions, such as drug rash with eosinophilia and systemic symptoms (DRESS), Stevens-Johnson syndrome, and toxic epidermal necrolysis, affect 1 to 10 in 10,000 new users of these antiseizure medications. Risk factors for cutaneous reactions include genetic predisposition, pediatric or old age, a previous history of cutaneous reactions with other medications, high initial dose and rapid escalation schedules, concomitant immune system disorders, and specific concurrent medications. Presence of the human leukocyte antigen HLA-B 1502 is strongly correlated with carbamazepine-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in patients with ancestry across broad areas of Asia, and evidence suggests these findings can also apply to phenytoin, lamotrigine, and oxcarbazepine. Among other HLA genotypes that predispose to antiseizure-induced reactions, the antigen HLA-A 3101 has been found to be associated with multiple carbamazepine-induced cutaneous reactions in Chinese, Japanese, and European populations.
Other serious idiosyncratic reactions to antiseizure medications include hepatotoxic effects with valproate, histologically evident as a microvesicular steatosis, in less than 1 in 20,000 patients, but this reaction is of particular concern in children under 3 years of age receiving multiple antiseizure medications (22). Felbamate usage is associated with aplastic anemia and hepatotoxicity in around 1 in 4000 patients. Rare cases of acute angle-closure glaucoma and myopia have been reported with topiramate (24).
Type C effects (long-term complications). Type C effects include chronic reactions related to cumulative medication exposure. They can be insidious because of their slow progressive development. Although some of these effects recede after discontinuation of the medication, others can be irreversible. Long-term complications of antiseizure medications tend to be medication specific. Chronic usage by many of the older agents can lead to dysmorphic changes, such as gum hypertrophy and hirsutism with phenytoin (15) and weight gain and polycystic ovarian syndrome with valproate (49). Weight gain is particularly associated with valproate, gabapentin, pregabalin, vigabatrin, and perampanel. Weight loss is associated with topiramate and zonisamide.
A serious type C effect is bilateral visual field loss induced by vigabatrin, which is irreversible and can worsen with continued treatment. Prevalence ranges from 14% to 92%. Suggested risk factors include male sex, older age, cumulative dose and mean daily dose of vigabatrin, and duration of exposure.
Treatment with topiramate and zonisamide carries a small risk of renal calculi via their shared carbonic anhydrase-inhibiting activity. This property may also account for their association with hypohidrosis, which can be symptomatic for patients living in tropical countries (08).
Long-term antiseizure medication therapy can produce hypocalcemia and decrease biologically active vitamin D levels, leading to reduced bone mineral density and a higher risk of fractures. Both enzyme-inducing and nonenzyme-inducing agents are implicated, and their effects may be additive. A variety of mechanisms have been suggested, the most significant of which is an increased rate of bone turnover. Patients at high risk of osteoporosis, eg, those on polytherapy or with poor mobility, are recommended to take calcium and vitamin D supplements and to have regular bone density measurements (01).
Psychiatric complications. Adverse psychiatric effects are noted in about 15% to 20% of patients with epilepsy who take antiseizure medications. These effects include behavioral or personality changes (irritability, hyperactivity, agitation, and aggressive behavior), depression, and psychosis. Some antiseizure medications, such as phenobarbital, topiramate, and vigabatrin are associated with neuropsychiatric complications and should be used cautiously in patients with a history of mental illness. Behavioral problems such as agitation, aggression, hostility, psychosis, anxiety, and depression have been reported in up to 7% of patients treated with levetiracetam, particularly those with a prior history of psychiatric illness (75). Zonisamide can also produce or exacerbate psychiatric comorbidities, particularly depression (18).
Furthermore, growing evidence suggests that individual vulnerability plays an important part. For example, family and personal psychiatric history, family history of epilepsy, personal history of febrile convulsions, and presence of tonic-atonic seizures are independent predictors for occurrence of psychiatric symptoms during treatment with topiramate. A positive psychiatric history seems to be an important risk factor for development of adverse psychiatric effects with other antiseizure medications (60). The United States Food and Drug Administration analyzed data from 199 placebo-controlled trials of 11 antiseizure medications and found an increase in risk of suicidal behavior or ideation among patients receiving antiseizure medications (0.43%) compared to those receiving placebo (0.22%) (US Food and Drug Administration 2008). There were four completed suicides among the 27,863 patients randomized to receive antiseizure medications compared with none among the 16,029 in the placebo groups. Although the difference was statistically significant, the absolute magnitude of the difference was small considering the sample size analyzed. Given that antiseizure medication therapy was evidently indicated for these patients, the implication of such findings on treatment alternatives is unclear. Whether other factors, such as indication of treatment- and medication-specific effects, may play a role is controversial. A subsequent large-scale community-based study found that the current use of antiseizure medications was not associated with an increased risk of suicide-related events among patients with epilepsy, but it was associated with an increased risk of such events among patients with depression and among those who did not have epilepsy, depression, or bipolar disorder (03). A community-based study of patients with treated epilepsy documented increased risk of suicidal behavior only in those receiving the newer antiseizure medications with a high risk of depression (levetiracetam, tiagabine, topiramate, vigabatrin) (02). These reports have raised renewed concern over adverse psychiatric effects of antiseizure medications in general, and it is advisable that patients receiving antiseizure medications should be monitored for the emergence or worsening of suicidal thoughts or behavior.
Type D effects. Teratogenic and carcinogenic effects are included in the type D category. It is generally accepted that antiseizure medication treatment during the first trimester of pregnancy is associated with a small, but significant, increase in the risk of major congenital malformations (defined as structural abnormalities with surgical, medical, or cosmetic importance). This risk is:
• probably no different or only slightly higher than the background (around 1% to 2%) in women with epilepsy who are not taking antiseizure medications. | |
• probably less than twice the background rate with commonly used antiseizure medications (other than valproate and topiramate) as monotherapy, although the relative risk may vary with individual antiseizure medications. | |
• certainly increased with valproate monotherapy to three to five times the background rate. | |
• likely to be high with topiramate, though the relevant studies are still inadequate. | |
• certainly higher with polytherapy than monotherapy; valproate is a significant contributor to the high risk of major congenital malformations in polytherapy, particularly in combination with lamotrigine (10%). | |
• likely to be dose-dependent, at least for valproate and probably for lamotrigine (ie, the higher the plasma antiseizure medication concentration, the higher the relative risk of major congenital malformations) (73). |
See details in articles on Pregnancy and epilepsy as well as in Fetal anticonvulsant syndrome.
Adverse cardiac effects of antiseizure medications. There is a conspicuous poverty of information and recommendations on the cardiac adverse medication reactions of antiseizure medications and their effect on the electrocardiogram. The situation may now change in view of the increasing number of studies on SUDEP (05) and the attention now given to cardiac function when assessing medication safety during the regulatory process, particularly with respect to ventricular repolarization as reflected in the prolongation of the ECG QT interval. Premarketing investigation of the safety of a new pharmaceutical agent now includes rigorous characterization of its effects on the QT/QTc interval labelled “Thorough QT/QTc Study” (57; 68). Antiseizure medications of sodium channel blockade contribute to ECG changes and probably SUDEP (05).
Carbamazepine |
Cardiac conduction disorders, hypertension or hypotension, bradycardia, arrhythmia, atrioventricular block with syncope, circulatory collapse, congestive heart failure, aggravation of coronary artery disease, thrombophlebitis, thrombo-embolism (eg, pulmonary embolism) |
Cenobamate |
Contraindicated with baseline short QT syndrome |
Eslicarbazepine acetate |
PR prolongation |
Everolimus |
Heart attack, arrythmias, heart failure, ejection fraction worsening |
Fenfluramine |
Changes in cardiac morphology with resultant left- and right-sided valvular regurgitation |
Gabapentin |
Palpitations |
Ganaxolone |
Tachycardia, syncope, cardiac arrhythmia |
Lacosamide |
PR prolongation |
Pregabalin |
Tachycardia, first degree heart block, sinus tachycardia, sinus arrhythmia, sinus bradycardia, congestive heart failure |
Rufinamide |
Shortening of the QT interval |
Zonisamide |
Shortening of the QT interval |
Cannabidiol |
None reported in product summaries |
Table 7 information presented as cited in the summaries of product characteristics; modified from (57).
Type E effects (adverse medication interactions). Adverse medication interactions are common with epilepsy treatment and are usually clinically relevant because:
• Most antiseizure medications have a narrow therapeutic index, and small changes in pharmacokinetics results in reduced efficacy or increased toxic effects. | |
• Many antiseizure medications affect the activity of medication-metabolizing enzymes. | |
• Most antiseizure medications are substrates of the same enzymes because these medications are typically taken for many years, the probability is high that at some point in life, people with epilepsy will be exposed to interactions with other medications used to treat intercurrent or concomitant disorders. | |
• Moreover, adverse medication interactions can arise when two or more antiseizure medications are co-prescribed (60). |
Long-term outcome. Long-term outcome studies suggest that 60% to 70% of newly diagnosed patients will become seizure-free with antiseizure medication therapy (40), mostly while taking their first or second antiseizure medication regimen (35; 52; 14). In line with this observation, the International League Against Epilepsy (ILAE) has proposed a consensus definition to define refractory or medication-resistant epilepsy as “a failure of adequate trials of two tolerated and appropriately chosen and used antiseizure medication schedules (whether as monotherapies or in combination) to achieve sustained seizure freedom” (34). It should be noted that the classification of a patient’s epilepsy as medication-resistant at a given point in time does not invariably imply that the patient will never become seizure-free on further antiseizure medication therapy (19; 44). Indeed, although approximately 60% of patients with newly diagnosed epilepsy appeared to have a constant course (either early remission or never seizure free), prognosis in the other 40% was less predictable, with some becoming seizure free after a delay and one in six demonstrating a “remitting-relapsing” course, fluctuating between periods of seizure freedom and recurrence (14). Thus, medication response can be a dynamic process. At least in the short term, prognosis for patients who do not respond to their first few medication regimens should not always be viewed nihilistically. Nonetheless, patients with epilepsy fulfilling the definition should be referred for a comprehensive review of the diagnosis and management, preferably at a specialist center. Workup for epilepsy surgery can be considered at this stage, particularly if a potentially operable structural abnormality, such as mesial temporal sclerosis, has been identified (17). If non-medication treatment options are not appropriate, further pharmacological modifications can still occasionally produce a worthwhile and sustained response.
Medication can be withdrawn slowly without seizure recurrence (at least over the following several years) in around 60% to 70% of adults who have been seizure-free for 2 years or more (47). Patients reporting few seizures before and after starting treatment, those taking a single medication, those seizure-free for many years, and those with no structural abnormality on brain imaging are likely to have the best outcomes. Myoclonic epilepsies carry a high risk of recurrence.
The value of the EEG in predicting relapse is small, particularly in adults (48). A normal EEG does not mean that antiseizure medication withdrawal is safe. Conversely, ictal EEG abnormalities associated with clinical manifestations (eg, jerks or absences) is a definite indicator for continuing proper antiseizure medication treatment.
The decision of whether to withdraw treatment should be made, whenever possible, by the patient, taking into account the likelihood of relapse and its potential psychosocial impact (eg, on driving and employment) balanced against the risk of adverse events from continued treatment (eg, teratogenicity) (57).
Consideration of total withdrawal of antiseizure medications is needed in the following patients:
• Patients who do not have epileptic seizures. | |
• Patients with agerelated and agelimited epileptic syndromes who have reached an appropriate age of remission. | |
• Patients who are seizure-free for more than 3 to 5 years, provided they do not have epileptic syndromes requiring longterm treatment, such as juvenile myoclonic epilepsy. |
Discontinuation of antiseizure medications should be extremely slow, in small doses and in long steps of weeks or months. Data in children suggest that a rate of 25% every 10 days to 2 weeks is safe (30). The rate of relapse increases with a faster rate of antiseizure medication discontinuation. Furthermore, with fast discontinuation of antiseizure medications, there is a risk of seizures that are directly related to the withdrawal effects of certain medications (phenobarbital and benzodiazepines). Before antiseizure medication withdrawal, there is a need for a thorough reevaluation of the patient, including an EEG in children (30). The presence of even minor and infrequent seizures specifies active disease. Conversely, the occurrence of such seizures in the process of medication discontinuation mandates restoration of antiseizure medication.
A significant report presents evidence-based nomograms to facilitate prediction of outcomes following medication withdrawal for the individual patient, including both the risk of relapse and the chance of long-term freedom from seizures (41). Independent predictors of seizure recurrence were epilepsy duration before remission, seizure-free interval before antiseizure medication withdrawal, age at onset of epilepsy, history of febrile seizures, number of seizures before remission, absence of a self-limiting epilepsy syndrome (childhood absence epilepsy, Rolandic epilepsy, or Panayiotopoulos syndrome), developmental delay, and epileptiform abnormality on EEG before withdrawal. Independent predictors of seizures in the last year of follow-up were epilepsy duration before remission, seizure-free interval before antiseizure medication withdrawal, number of antiseizure medications before withdrawal, female sex, family history of epilepsy, number of seizures before remission, focal seizures, and epileptiform abnormality on EEG before withdrawal (41).
Over-medication in terms of the number of antiseizure medications and doses and length of exposure is undesirable but common. Treatment should be reviewed at regular intervals to ensure that patients are not maintained for long periods on medications that are ineffective, poorly tolerated, or not needed and that concordance with prescribed medication is maintained.
Although it would be ideal for a woman contemplating pregnancy to have antiseizure medication treatment withdrawn, for many this would result in recurrence or exacerbation of seizures, which can be dangerous for both mother and baby. If the criteria for discontinuation are met, this should be done prior to conception in a planned pregnancy counseling program. If antiseizure medications cannot be withdrawn completely, treatment should be tapered to a minimally effective dose of, if possible, a single medication. See details in the article on Pregnancy and epilepsy.
Epileptic seizures result from abnormal, excessive, and synchronous excitation of neuronal populations. Antiseizure medications suppress their occurrence by modifying the bursting properties of neurons and by reducing hypersynchronization, thereby inhibiting the spread of abnormal neuronal firing (64). Although an antiepileptogenic effect has been noted for some antiseizure medications in selected animal models (66), such a property has not been demonstrated in clinical trials (72).
The mechanisms of action of the currently marketed antiseizure medications involve alterations in the balance between neuronal excitation and inhibition (Table 8). Three basic mechanisms are recognized: (1) modulation of voltage-gated ion channels (including sodium, calcium, and potassium channels), (2) enhancement of synaptic inhibitory neurotransmission, and (3) attenuation of brain excitation (Table 8). Many antiseizure medications have multiple modes of action, whereas the mechanisms for others are still unclear.
Blocking voltage-dependent Na+ channels (decreased Na+) | |
• Carbamazepine | |
Multiple, mainly or including blocking voltage-dependent Na+ channels | |
• Phenobarbital (decreased Na+, decreased Ca2+, increased GABA, decreased glutamate) | |
Increasing GABA inhibition (increased GABA) | |
• Clobazam (GABAA) | |
Blocking T-type Ca2+ channels (decreased Ca2+) | |
• Ethosuximide | |
Modifying Ca2+ channels and neurotransmitter release | |
• Gabapentin | |
Novel | |
• Brivaracetam: probably by synaptic-to-synaptic vesicle protein SV2A but still uncertain |
Table 8 information based on data from summaries of product characteristics and product inserts; modified from (57).
Voltage-gated ion channels regulate the electrical behavior of neurons as well as the release of neurotransmitters. Medications such as phenytoin, carbamazepine, and lamotrigine act primarily by limiting sustained, repetitive firing via blockade of voltage-gated sodium channels. This property is shared by some of the newer medications, such as oxcarbazepine, zonisamide, and eslicarbazepine. Gabapentin and pregabalin exert their antiseizure and other pharmacological effects by binding to the alpha2-delta subunit of the calcium channel.
Several antiseizure medications act primarily by enhancing GABAergic inhibition. The barbiturates and benzodiazepines modulate GABAA receptors. Vigabatrin and tiagabine inhibit the enzymatic degradation and reuptake of GABA, respectively.
Levetiracetam (binding to synaptic vesicle protein SV2A), lacosamide (selectively enhancing slow inactivation voltage-gated sodium channels), and perampanel (selective, noncompetitive, AMPA receptor antagonist) have novel modes of action.
All contributors' financial relationships have been reviewed and mitigated to ensure that this and every other article is free from commercial bias.
David Gloss MD
Dr. Gloss of The NeuroMedical Center in Baton Rouge has no relevant financial relationships to disclose.
See ProfileAppaji Rayi MBBS
Dr. Rayi of the Charleston Area Medical Center has no relevant financial relationships to disclose.
See ProfileJohn M Stern MD
Dr. Stern, Director of the Epilepsy Clinical Program at the University of California in Los Angeles, received honorariums from Ceribell, Jazz, LivaNova, Neurelis, SK Life Sciences, Sunovian, and UCB Pharma as advisor and/or lecturer.
See ProfileNearly 3,000 illustrations, including video clips of neurologic disorders.
Every article is reviewed by our esteemed Editorial Board for accuracy and currency.
Full spectrum of neurology in 1,200 comprehensive articles.
Listen to MedLink on the go with Audio versions of each article.
MedLink®, LLC
3525 Del Mar Heights Rd, Ste 304
San Diego, CA 92130-2122
Toll Free (U.S. + Canada): 800-452-2400
US Number: +1-619-640-4660
Support: service@medlink.com
Editor: editor@medlink.com
ISSN: 2831-9125
Neuro-Oncology
Oct. 03, 2024
Epilepsy & Seizures
Oct. 02, 2024
Epilepsy & Seizures
Sep. 16, 2024
Epilepsy & Seizures
Sep. 06, 2024
Epilepsy & Seizures
Sep. 06, 2024
Sleep Disorders
Sep. 05, 2024
Epilepsy & Seizures
Aug. 26, 2024
Epilepsy & Seizures
Aug. 23, 2024