Você está na página 1de 11

Epilepsia, 44(Suppl.

11):27–37, 2003
Blackwell Publishing, Inc.
C International League Against Epilepsy
!

Treatment Strategies for Myoclonic Seizures and Epilepsy


Syndromes with Myoclonic Seizures

∗ James W. Wheless and †Raman Sankar

∗ Department of Neurology and Pediatrics, Texas Comprehensive Epilepsy Program,University of Texas – Houston, Houston, Texas;
and †Department of Neurology and Pediatrics, Mattel Children’s Hospital at UCLA,University of California – Los Angeles,
Los Angeles, California, U.S.A.

Summary: Despite the availability of numerous treatment op- ered when making decisions regarding treatment. Rarely, some
tions, the diagnosis and treatment of myoclonic seizures con- antiepileptic drugs may exacerbate myoclonic seizures. Most
tinue to be challenging. Based on clinical experience, valproate epileptic myoclonus can be treated pharmacologically, but some
and benzodiazepines have historically been used to treat my- cases respond better to surgery, the ketogenic diet, or vagus nerve
oclonic seizures. However, many more treatment options exist stimulation. Because myoclonic seizures can be difficult to treat,
today, and the clinician must match the appropriate treatment clinicians should be flexible in their approach and tailor therapy
with the patient’s epilepsy syndrome and its underlying etiol- to each patient. Key Words: Myoclonic seizures—Epilepsy—
ogy. Comorbidities and other medications must also be consid- Antiepileptic drug therapy.

Single myoclonic jerks in patients with epilepsy have oclonic seizures. If no reversible etiology is found, the
long been recognized. Myoclonus comes from the Greek myoclonus, which is one component of an epilepsy syn-
myo, meaning muscle, and klonus meaning turmoil. While drome, can be treated along with the syndrome. Accurate
this term was used by the Greeks to describe the erratic definition of a patient’s epilepsy syndrome is an important
movement often seen with myoclonic jerks, it aptly de- factor in treatment. Comorbidities and other medications
scribes the current state of therapy for myoclonic seizures must also be considered when making decisions regarding
and myoclonic epilepsies. Jeavons addressed the prob- treatment.
lem of nosology of the myoclonic epilepsies in 1977 (1), Historically, valproate (VPA) and clonazepam (CZP)
suggesting that myoclonic epilepsies were as difficult to have been used to treat myoclonic seizures based on
classify as they were to treat. Myoclonus has become a de- clinical experience. Both drugs work by promoting γ -
scription for many semiologically and nosologically dif- aminobutyric acid (GABA) action in the brain. However,
ferent conditions. The treatment of myoclonic epilepsies many more options are available today, and clinicians
is still problematic 25 years later (2–7). must match the appropriate treatment with the epilepsy
One task for clinicians is to establish whether or not syndrome and its underlying etiology. Most epileptic my-
the paroxysmal episodes that the patient is experiencing oclonus is treated medically, but some cases may respond
constitute epilepsy. The first step in this process is to to surgery, the ketogenic diet, or vagus nerve stimulation.
determine if a reversible etiology is present. The physi- Animal models of myoclonic seizures have played a
cian is then confronted with a clinical conundrum. My- role in evaluating the possible efficacy of drug therapy for
oclonus may only be one part of an epilepsy syndrome, treatment of human myoclonic seizures. Animal models
and several problems regarding its treatment exist: (a) not can be classified as those involving provoked seizures and
all antimyoclonic drugs are antiepileptic, (b) only some those involving genetic models. Provocation of seizures
antiepilepsy drugs (AEDs) are antimyoclonic, (c) many in the former is commonly accomplished by chemical
myoclonic epilepsies are refractory to treatment, and or electrical means, although other means, such as hy-
(d) some AEDs may exacerbate myoclonus or induce my- poxia and intermittent light stimulation, have also been
used. Classic pharmacologic screening involving rodents,
as used by the Anticonvulsant Screening Project of the
Address correspondence and reprint requests to Dr. J. W. Wheless at National Institute of Neurologic Disorders and Stroke, has
Texas Comprehensive Epilepsy Program, University of Texas – Hous-
ton, 6431 Fannin St., Suite 7044, Houston, TX 77030, U.S.A. E-mail: produced inconsistent information for predicting the effi-
James.W.Wheless@uth.tmc.edu cacy of potential therapeutic agents against myoclonus.

27
28 J. W. WHELESS AND R. SANKAR

With earlier-generation AEDs, models were only some-


what predictive of efficacy. Only CZP and VPA have
demonstrated efficacy in animal models for protection
against all three seizure provocations involving subcuta-
neous pentylenetetrazol, bicuculline, and picrotoxin (8).
Ethosuximide (ESM), which was only marginally ac-
tive against bicuculline, has been shown not to be a
useful antimyoclonic AED (8,9). Phenytoin (PHT) and
carbamazepine (CBZ) clearly distinguished themselves
as agents that were mainly active in the maximal elec-
troshock model (10). Among new-generation AEDs, fel-
bamate (FBM) has shown clinical utility in a variety
of myoclonic seizures and was also active in protect-
ing rats against subcutaneous pentylenetetrazol, subcu-
taneous bicuculline, and subcutaneous picrotoxin (11).
Gabapentin (GBP), the next agent to arrive on the United
States market, has been shown to be ineffective in sub-
cutaneous bicuculline and picrotoxin provocations in the
rodent (12) and appears to be clinically useful only for par-
tial and secondarily generalized seizures (13). Vigabatrin
(VGB), like GBP, has not demonstrated significant activity
against subcutaneous bicuculline or picrotoxin challenge,
and is not a clinically useful agent for most syndromes
involving myoclonic seizures (14). Experience with GBP
highlights another difficulty in the use of animal mod-
els for screening compounds to treat myoclonus. Kan-
FIG. 1. A: Baboon in the primate chair for EEG recording.
thasamy et al. (15) found GBP to be antimyoclonic in a B: EEG demonstrating a photomyoclonic response in Papio
posthypoxic rodent model, but not for myoclonus induced hamadryas anubis, cynocephalus, an animal model for photosen-
by administration of p,p# -DDT (1,1,1-trichloro-2,2-bis(p- sitive epilepsy. (Figure courtesy of Dr. C. Akos Szabo, University of
Texas Health Sciences Center, San Antonio, TX, U.S.A., reprinted
chlorophenyl)ethane). with permission.)
Experience with other new-generation AEDs has also
revealed many inconsistencies. Lamotrigine (LTG), which
has an animal testing profile resembling that of PHT (16), in the baboon (25), suggesting that the baboon model also
has demonstrated clinical utility for treating some patients has limitations in its ability to predict the clinical utility
with juvenile myoclonic epilepsy (JME) (17). Paradox- of test AEDs. This model is apparently not routinely used
ically, LTG is also known to exacerbate myoclonus in in screening, and many of the new-generation AEDs have
some patients (18–20). Topiramate (TPM), another new- no published literature describing their use in this model.
generation agent, has been shown to be ineffective in Another well-characterized genetic model involves the
blocking chemically induced seizures (21). Nevertheless, tottering mouse. The tg locus causes a delayed-onset re-
this medication may be of value in treating a variety of cessive neurological disorder in the mouse, featuring a
syndromes that involve myoclonus, as is discussed later stereotyped triad of ataxia, intermittent focal myoclonic
in this article. seizures, and bursts of generalized 6- to 7-Hz spike waves.
The most extensively described genetic model of This model has not been routinely used in screening for
epilepsy involves Papio papio, the photosensitive baboon AEDs; hence, its reliability for this purpose is unknown.
(Fig. 1A). Killiam et al. (22) first reported that when these Other genetic models of inherited myoclonus include a
animals were subjected to intermittent light stimulation, disorder of the inhibitory glycine receptor that results in
paroxysmal discharges in the form of spikes and waves spontaneous and stimulus-sensitive myoclonus in Polled
or polyspikes and waves resulted (Fig. 1B). Behaviorally, Hereford calves (26) and spontaneous and photically in-
bilateral and synchronous myoclonic jerks appeared and duced myoclonus in a mutant strain of Fayoumi chickens
were followed by generalized convulsive seizures. The in- (27). The possible importance of the glycine receptor in
volvement of the GABAergic system in this model was de- some myoclonic disorders is highlighted by the recent de-
scribed in detail by Meldrum and Wilkins (23). This model velopment of a transgenic mouse model of hyperekplexia,
correctly predicted GBP’s lack of efficacy in human my- which involves a mutation in the gene for the α 1 sub-
oclonic epilepsies (24). However, VGB was shown to be unit of the glycine receptor (28). However, the cost and
effective in blocking photically induced epileptic activity inconsistent availability of these animals precludes their

Epilepsia, Vol. 44, Suppl. 11, 2003


MYOCLONUS TREATMENT STRATEGIES 29

routine use for developing highly specific antimyoclonic demonstrated some efficacy, response to treatment is typ-
drugs. Economic forces continue to drive the development ically poor and seizure freedom is rarely achieved. None
of AEDs for partial seizures, with subsequent adoption of of the conventional AEDs, adrenocorticotropic hormone
some AEDs to treat myoclonic seizures based on clinical (ACTH) gel, prednisone, or pyridoxine has been effec-
experience. tive in treating this syndrome (30). The myoclonias of this
No controlled clinical trials have solely evaluated the syndrome gradually decrease with age.
efficacy of treatment for myoclonic seizures; rather, they The treatment of infantile spasms is based on the under-
have evaluated the effect of treatment on epilepsy syn- lying etiology of the spasms (31,32). With respect to both
dromes that may include myoclonus. This practice has seizure control and developmental outcome, infants who
resulted from the difficulty in accurately quantifying my- have underlying focal cortical dysplasia, porencephalic
oclonic seizures before and after treatment in controlled cysts, or (rarely) a brain tumor may show the best re-
clinical trials. This limitation must be acknowledged when sponse to epilepsy surgery. Anecdotal and controlled trial
evaluating drug treatment strategies for myoclonic epilep- data indicate that VGB is the treatment of choice for in-
sies. In this review of treatment options, the American fantile spasms associated with tuberous sclerosis complex
Academy of Neurology Quality Standards Subcommit- (33–37). Children with cryptogenic infantile spasms show
tee practice guidelines (29) are used to classify the levels the best response to ACTH in clinical trials (32,38). Use
of evidence for each treatment. These recommendations of VGB (32–35,39) and nitrazepam (NTZ) (40) to treat
are based on the following levels of evidence for thera- cryptogenic infantile spasms has also been evaluated in
peutic modalities: (a) class I—controlled clinical trials; clinical trials. In Japan, pyridoxine is typically admin-
(b) class II—case-controlled and cohort studies; and (c) istered as initial therapy for infantile spasms but has a
class III—evidence provided by case series, case reports, low response rate (41–43). Other treatment options that
and expert opinions. For most epilepsy syndromes with have been pursued in case series include VPA, prednisone,
myoclonic seizures, the controlled trial data (class I evi- FBM, LTG, TPM, and zonisamide (ZNS) (38,39,44,45).
dence) only applies to the overall treatment of the epilepsy The ketogenic diet has recently shown success in children
syndrome, not specifically to the myoclonic component. with refractory infantile spasms (cryptogenic and symp-
As a result, most information presented in this article is tomatic etiologies) (46). Children with symptomatic eti-
based on class II or class III evidence for the treatment of ologies are often treated with the same medications, but the
myoclonic seizures. Additionally, all the information pre- response rate is much lower than in children with a cryp-
sented herein regarding therapies that may cause exacer- togenic etiology (32,38,39,44). As such, in patients with
bation of myoclonic seizures is based on class III evidence symptomatic etiologies, a risk/benefit assessment must be
(typically case series). made for each therapeutic option and discussed with the
child’s parents.
Severe myoclonic epilepsy in infancy (Dravet syn-
EPILEPSY SYNDROMES
drome) begins with partial or generalized, often pro-
WITH MYOCLONIC SEIZURES
longed, febrile seizures in normal infants before 1 year
Syndromes of infancy and early childhood of age. Myoclonus occurs between the ages of 1 and 4
Myoclonic seizures that begin in infancy are typi- years, typically upon awakening. Children progress to ex-
cally associated with both poor response to treatment and perience multiple seizure types, and their psychomotor de-
poor ultimate prognosis (3). Included in this category are velopment becomes retarded after the second year of life.
early myoclonic encephalopathy (neonatal myoclonic en- Photoparoxysmal effects are common in this syndrome.
cephalopathy), infantile spasms, and severe myoclonic Recent placebo-controlled, multicenter studies have eval-
epilepsy in infancy (Dravet syndrome). Benign myoclonic uated the efficacy of stiripentol in the treatment of severe
epilepsy of infancy is an exception in that it can have a myoclonic epilepsy in infancy (Dravet syndrome) (47,48).
good response to treatment and a favorable prognosis. For In one study by Chiron et al. (47), children were typically
infants and young children, there are few conditions that treated with VPA and clobazam during the baseline phase
fall between these two ends of the spectrum. (Fig. 2). They were then randomized to treatment with
Massive or axial bilateral myoclonias are the myoclonic
features of early myoclonic encephalopathy. This rare syn-
Stiripentol (n=21)
drome has onset during the first month of life and is often
fatal by 1 year of age. The electroencephalogram (EEG)
Valproate, clobazam Stiripentol
in patients with this condition reveals a burst-suppression
pattern and may evolve toward typical hypsarrhythmia. No 0 to 4 weeks 50 to 100 mg/kg/d
Baseline
controlled clinical trials have investigated the treatment of Placebo (n=20)
early myoclonic encephalopathy. Treatment options are all 8 weeks

based on anecdotal reports, and although drug therapy has FIG. 2. Severe myoclonic epilepsy in infancy (47) study design.

Epilepsia, Vol. 44, Suppl. 11, 2003


30 J. W. WHELESS AND R. SANKAR

stiripentol (STP) or placebo for 8 weeks. Subsequently, spectrum AEDs with demonstrated efficacy in the treat-
patients entered an open-label phase, during which they ment of Lennox-Gastaut syndrome in controlled clinical
were treated with STP at dosages of 50–100 mg/kg/day. trials (57–63). The availability of these two drugs has been
Responder rates (i.e., proportions of patients experienc- a major asset to pediatric neurologists who previously had
ing a >50% decrease in seizure frequency) were sig- only VPA available. Additionally, there have been anec-
nificantly greater for the STP group (71%) than for the dotal reports of success in treating Lennox-Gastaut syn-
placebo group (5%). The proportion of patients who were drome with ZNS, steroids, CZP, MSM, and NTZ (51).
free from generalized clonic and generalized tonic-clonic Nonpharmacologic treatments can also be of benefit in
seizures during the study was also significantly greater treating Lennox-Gastaut syndrome. Both the ketogenic
in the STP group (43%) than in the placebo group (0%). diet and vagus nerve stimulation may be particularly use-
Aside from documenting the efficacy of STP in this very ful in the treatment of drop attacks associated with this
difficult-to-treat childhood epilepsy syndrome, this study condition (64–69). Vagus nerve stimulation has been so
demonstrated the use of a multicenter group to determine efficacious in the treatment of drop attacks that corpus
a drug’s efficacy in a relatively uncommon epilepsy syn- callosotomies are seldom performed. However, when such
drome, for which recruiting enough patients at a single procedures are performed, there appears to be a synergis-
center to complete a trial would likely be impossible. Be- tic effect in patients who had previously received vagus
fore this trial, treatment of severe myoclonic epilepsy in nerve stimulation (70). Interestingly, GABAergic drugs,
infancy was based on anecdotal evidence for the efficacy such as VGB and GBP, may exacerbate the myoclonic
of drugs, such as VPA, CZP, methsuximide (MSM), TPM, seizures specifically associated with Lennox-Gastaut syn-
and intravenous gammaglobulin. Both LTG and CZP are drome (71,72). Epileptic myoclonus in Lennox-Gastaut
reported to exacerbate seizures in this condition (18). syndrome originates from a stable generator in the frontal
Benign myoclonic epilepsy of infancy is the only cortex and spreads to other cortical areas, whereas the
epilepsy syndrome associated with myoclonic seizures in myoclonus in Doose syndrome appears to be a primary
infancy or childhood that can have a favorable prognosis. generalized epileptic phenomenon (73).
Seizures are characterized by symmetric myoclonus, and Doose syndrome (myoclonic-astatic epilepsy of early
this is the only seizure type associated with this syndrome childhood) occurs in children between 2 and 5 years of
(with the exception of occasional febrile convulsions). age. The jerks associated with this syndrome, followed
Generalized tonic-clonic seizures may develop later dur- by atonia, often lead to abrupt falling and injury. No for-
ing adolescence. The EEG background for individuals mal clinical trials have documented the efficacy of AEDs
with this syndrome is normal, and the myoclonus is ac- in treating Doose syndrome, and information regarding
companied by a generalized spike or polyspike and slow- its treatment is based on anecdotal experience. FBM ap-
wave complex. There have been no controlled trials to pears to play a special role in the treatment of Doose syn-
evaluate treatment options for this syndrome, but anec- drome, as many patients with this condition are uniquely
dotal evidence suggests that VPA, TPM, LTG, and CZP responsive to the drug. Other AEDs that have been used
are effective (49). Some children may require relatively in treating Doose syndrome include ESM, ACTH, VPA,
brief therapy (i.e., 1–2 years), while others may require CZP, TPM, and LTG (2,74–76). Additionally, the keto-
longer-term therapy. genic diet and vagus nerve stimulation have been used as
The ability of a drug to effectively treat Lennox-Gastaut nonpharmacologic agents for treatment of intractable drop
syndrome has often been considered the gold standard attacks sometimes associated with this condition. Exacer-
for declaring an AED to have broad-spectrum efficacy in bation of myoclonic astatic seizures has been reported in
childhood epilepsy. VPA has been used to treat Lennox- patients treated with PHT, CBZ, and VGB (77).
Gastaut syndrome for 25 years, and there is a wealth of
clinical evidence indicating its efficacy in open-label tri- Syndromes of late childhood, adolescence,
als, although no double-blind trials have been performed and adulthood
(31,50–54). It was not until ∼15 years after the intro- Three of the late-childhood and adolescent-onset ab-
duction of VPA in the United States that a successful sence epilepsy syndromes may be associated with my-
controlled clinical trial was conducted to investigate the oclonic seizures. Juvenile absence epilepsy is typically
treatment of Lennox-Gastaut syndrome; FBM was the treated with VPA, but LTG and CZP also appear to be
first drug to show efficacy in this manner (55,56). Un- efficacious (74,78–81). TPM is probably less efficacious
fortunately, potential side effects of FBM have limited in the treatment of absence seizures than either LTG or
its use in Lennox-Gastaut syndrome, but the drug is still VPA (61,82). As such, if absence seizures are a major
recommended as a third- or fourth-line agent. FBM is component of the patient’s childhood epilepsy syndrome,
particularly efficacious in controlling the drop attacks the situation warrants use of LTG over that of TPM. The
associated with Lennox-Gastaut syndrome. Two other GABAergic agents VGB and tiagabine have also been re-
drugs (i.e., LTG and TPM) have also emerged as broad- ported to exacerbate absence seizures associated with this

Epilepsia, Vol. 44, Suppl. 11, 2003


MYOCLONUS TREATMENT STRATEGIES 31

condition (74,77). Tassinari syndrome (epilepsy with my- simple tremor, may be considered a side effect of VPA
oclonic absences) has been treated with VPA, CZP, LTG, therapy.
TPM, ESM, and phenobarbital (PB) (2,83). A similar FAME is characterized by adult-onset myoclonus in
condition, Jeavons syndrome (eyelid myoclonia with ab- the extremities (cortical tremor), generalized myoclonic
sences), has been treated with VPA, CZP, ESM, and LTG jerks, rare generalized tonic-clonic seizures, generalized
(2,84). The last two are uncommon epilepsy syndromes; EEG abnormalities, and normal IQ (95,96). A locus for
therefore, no clinical trials have investigated treatment for this form of epilepsy has been mapped to chromosome
them. 8q24 in Japanese families (95,96), although a European
JME is the most common adolescent primary gener- pedigree did not have this linkage (97), suggesting ge-
alized epilepsy syndrome. Approximately 18 years ago, netic heterogeneity of this condition. Clinical and EEG
the unique sensitivity of this syndrome to treatment with characteristics suggest that FAME is a form of idiopathic
VPA was first proposed (85). Since then, although no for- generalized epilepsy akin to JME (95–97). CZP was ef-
mal clinical trials have been conducted, there have been fective in all treated FAME patients (97).
numerous open-label reports documenting VPA’s clinical The progressive myoclonic epilepsies (PMEs) are a
efficacy in this condition (86–88). Only one controlled challenging group of disorders, both from a treatment and
clinical trial has investigated the treatment of JME. This a diagnostic standpoint (98,99). The treatments used in
trial evaluated the efficacy of adjunctive therapy with TPM these conditions often are symptomatic and may improve
in patients with primary generalized tonic-clonic seizures, seizure control or myoclonus, allowing the patient greater
a subgroup of which had JME (61,89). In the TPM-treated mobility or quality of life. However, such treatments do
JME patients, primary generalized tonic-clonic seizures not reverse the underlying etiology and the progressive
were reduced >50% in 73% of patients, versus 18% of encephalopathy associated with these conditions. Despite
placebo-treated patients (p = 0.03). Open-label experi- this major limitation, treatments are often very benefi-
ence in the treatment of JME with LTG, ZNS, FBM, and cial to the patient and the family, as they allow patients
CZP has also been reported (2,7). Older AEDs, such as to be mobile for as long as possible and decrease the
primidone, PB, acetazolamide (ACZ), MSM, and ESM, number of injuries secondary to myoclonic seizures. No
were also used to treat JME before the introduction of formal clinical trials have been conducted in the treat-
VPA. Reports indicate that ACZ, though at times effective ment of PME; however, open-label studies and anecdo-
for controlling generalized tonic-clonic seizures, is rarely tal reports provide evidence for the usefulness of some
effective for controlling myoclonic seizures (90). Benzo- drugs. Open-label clinical trials have reported the effi-
diazepines (BZDs) may be very effective in controlling the cacy of ZNS in the treatment of PMEs (100–102). In the
myoclonus associated with JME (88); however, this may treatment of myoclonus epilepsy with ragged-red fibers
deprive the patient of the warning jerks before the onset (MERRF), both carnitine and coenzyme Q10 may help
of a generalized tonic-clonic seizure. PHT, CBZ, LTG, improve seizure control (103). LTG has reported special
and VGB have all been associated with exacerbation of efficacy in the treatment of neuronal ceroid lipofuscinosis
myoclonic seizures in JME (34,77,91–93). (104,105). Other medical therapies that have been used
Guerrini et al. recently proposed a new epilepsy syn- in the treatment of PMEs include piracetam, levetirac-
drome linked to chromosome 2p11.1–q12.2, which they etam (LEV), CZP, and VPA (106,107). Historically, both
defined as autosomal dominant cortical myoclonus and alcohol and N-acetylcysteine have been helpful in some
epilepsy (ADCME) (94). Eight individuals from a single patients with PMEs (108,109). However, a recent report
pedigree presented with a nonprogressive disorder with has shown variable response and some notable side effects
onset between the ages of 12 and 50 years. Most indi- during treatment with N-acetylcysteine and has suggested
viduals had rare seizures. The disorder is characterized that a multicenter, controlled trial be performed (110).
by predominantly distal semicontinuous rhythmic my- Single case reports indicate that vagus nerve stimulation
oclonus and generalized tonic-clonic seizures (all patients) can improve seizures in patients with PME (111). PHT
and complex partial seizures (three patients). Myoclonic has been reported to exacerbate the seizures specifically
seizures are not a prominent feature. This syndrome has associated with Unverricht-Lundborg disease (112); this
some clinical and neurophysiological characteristics that effect may possibly occur in other PMEs, although it has
are similar to familial adult myoclonic epilepsy (FAME), not been reported.
including generalized EEG abnormalities. All patients The recently described epilepsy syndrome, generalized
treated with CBZ had severe worsening of their my- epilepsy with febrile seizures plus (GEFS+), has a pheno-
oclonus. CZP, VPA, and PB, drugs combining antiepilep- type with a significant component of myoclonic seizures
tic and antimyoclonic properties, produced the most ben- (113,114). No formal clinical trials have been conducted,
efit. Patients with this syndrome may be misdiagnosed as but anecdotal evidence suggests efficacy of VPA and
having JME, and the cortical tremor, if interpreted as a BZDs for the treatment of this condition (113,114).

Epilepsia, Vol. 44, Suppl. 11, 2003


32 J. W. WHELESS AND R. SANKAR

SPECIAL SITUATIONS Postanoxic myoclonus has historically been treated


with VPA or BZDs. No controlled clinical trials have been
Chromosomal encephalopathies, metabolic disorders,
conducted in the treatment of this condition; however, re-
and anoxic insults may be associated with refractory my-
cent clinical reports have suggested good efficacy of LEV
oclonic seizures. Down’s syndrome (trisomy 21) may
(125–127).
present in infancy or early childhood with myoclonic
seizures or infantile spasms. Typically, the seizures show a
good response to treatment with AEDs, steroids, or ACTH AED THERAPY FOR MYOCLONIC SEIZURES
(44,115–118). Late-onset myoclonic epilepsy in Down’s The administration of VPA as one of the primary treat-
syndrome should be considered in the differential diag- ments for myoclonic seizures is based on a long history of
nosis of adult-onset PMEs (119). Wolf-Hirschhorn syn- clinical experience (Fig. 3) (1,53). Although VPA is gen-
drome (partial monosomy 4p or 4p-) is a chromosomal erally regarded as the drug of choice for Lennox-Gastaut
defect that may affect the alpha-2 and the beta-1 genes for syndrome, seizures are effectively controlled in only 10–
the GABAA receptor (120). Reduction in the amount of 30% of patients (1). VPA has also been used to treat
gene product could contribute to the high epileptogenic- Doose syndrome, myoclonic absence, photomyoclonic
ity of this syndrome. Wolf-Hirschhorn syndrome shows epilepsy, eyelid myoclonia with absence seizures, JME,
some similarities with the electroclinical findings of An- and postanoxic myoclonus (49,128,129). The availability
gelman syndrome. As such, GABAergic drugs (VPA and of an intravenous formulation of VPA allows rapid loading
BZDs) usually control the seizures, while CBZ may exac- for treatment of myoclonus status epilepticus associated
erbate the seizures in this syndrome (120,121). Angelman with any of these epilepsy conditions (130). Before intra-
syndrome (partial monosomy 15Q) is another chromo- venous VPA was available, the only intravenous formula-
somal defect that affects the GABA receptor by elimi- tion available for the treatment of myoclonic epilepsy was
nating a cluster of GABAA receptor genes. The resultant a BZD.
decreased inhibition has been suggested to lead to cortical LTG was introduced as a second broad-spectrum AED,
hyperexcitability (121). BZDs, VPA, and TPM may con- and controlled clinical trials have demonstrated the drug’s
trol the seizures associated with Angelman syndrome, and efficacy for myoclonic epilepsy as part of the treatment
piracetam has been reported to decrease the tremulous- for Lennox-Gastaut syndrome (57). In addition, anecdo-
ness (probable cortical myoclonus) associated with this tal reports have shown the efficacy of LTG in infantile
gene abnormality (122). Again, CBZ has been reported to spasms, Doose syndrome, seizures associated with Rett
worsen the myoclonic seizures in this syndrome (121). syndrome, Jeavons syndrome, JME, Tassinari syndrome,
Two other chromosomal abnormalities have been asso- juvenile absence epilepsy, photosensitive epilepsy, and
ciated with myoclonic seizures, although no specific ther- neuronal ceroid lipofuscinosis (17,81,131–134). A con-
apeutic approaches have been reported. In trisomy 1p-, trolled double-blind trial of LTG in JME is currently
three voltage-gated potassium channel genes are clustered being conducted. Interestingly, the combination of VPA
together in the 12p13 band. TPM is the only AED that is with LTG appears to be synergistic (135), making this
known to affect the potassium receptor and potentially a useful combination for epilepsy syndromes associated
could show efficacy in this condition. Patients with an with refractory seizures. However, one study has shown
inverted duplicated chromosome 15 have three GABAA that LTG may aggravate severe myoclonic epilepsy in
receptor subunit genes that may be altered. Although no infancy (18).
drugs have specifically been tested, GABAergic agents TPM has been extensively evaluated in treating many
may have potential efficacy in this condition. epilepsy syndromes (59–62,89,122,136,137). Controlled,
Several metabolic disorders are associated with my-
oclonic seizures. Some of these disorders, such as
pyridoxine-dependent epilepsy and biotin deficiency, may 100
Percentage of Patients Who

require life-long treatment with the necessary cofactor 71%


70%
(42,43). Other disorders may respond to specific dietary
Were Seizure Free

53%
manipulation. Glucose transporter protein deficiency re- 50
sponds uniquely to the ketogenic diet, and this diet must
18%
be maintained throughout the patient’s life (123,124). Spe-
cific dietary treatment is also used for treatment of maple 0
0%

syrup urine disease. Unfortunately, peroxisomal disorders, Myoclonic LGS (n=38) Jeavons JME (n=23) BMEI
Absence Syndrome (n=17)
molybdenum cofactor deficiency, and nonketotic hyper- (n=6) (n=19)
glycinemia segmental myoclonic jerks all must be treated
FIG. 3. Treatment of myoclonic seizures: valproate monother-
symptomatically because no other therapies are available apy (53). JME, juvenile myoclonic epilepsy; BMEI, benign my-
at this time. oclonic epilepsy of infancy; LGS, Lennox-Gastaut syndrome.

Epilepsia, Vol. 44, Suppl. 11, 2003


MYOCLONUS TREATMENT STRATEGIES 33

100 NONPHARMACOLOGIC THERAPY


≤ 50% 100% FOR MYOCLONIC SEIZURES
68%
Any type of focal cortical lesion can cause focal cor-
Patients (%)

53%
50 tical myoclonus (e.g., angiomas, tumor, or encephalitis).
Rarely, focal cortical dysplasia may be associated with
14% cortical myoclonus. Surgical excision of the excitable
9%
tissue can cure the myoclonus. Patients with congenital
0
GTC Myoclonic GTC Myoclonic
hemiplegia due to vascular causes or congenital brain mal-
(n=34) (n=36) (n=34) (n=36) formations may show asymmetric myoclonic seizures as
FIG. 4. Treatment of Lennox-Gastaut syndrome with open-label part of their epilepsy syndrome. Successful hemispherec-
topiramate (59): seizure reduction for ≥6 months. GTC, general- tomy resolves the myoclonus along with the other seizure
ized tonic-clonic. types. Epilepsy partialis continua is considered a par-
ticularly treatment-resistant form of cortical myoclonus
double-blind trials have demonstrated TPM’s efficacy in but may be alleviated by functional hemispherectomy
the treatment of Lennox-Gastaut syndrome (59,62) and in patients with Rasmussen’s encephalitis. Patients with
primary generalized tonic-clonic seizures, including JME hypothalamic hamartomas may display generalized my-
(Figs. 4 and 5) (61,89). Efficacy of TPM as add-on open- oclonic seizures as part of their epilepsy syndrome. The
label therapy has been shown in the treatment of severe ketogenic diet, vagus nerve stimulation, surgical resec-
myoclonic epilepsy in infancy and other childhood my- tion, or gamma knife treatment may improve the seizures
oclonic epilepsies (60,122,136,137). (152–156). The more complete the resection, the better
ZNS has multiple mechanisms of action, suggesting the result.
it may also be a broad-spectrum AED. Anecdotal re- Vagus nerve stimulation has also been reported to im-
ports suggest efficacy of ZNS in treating Lennox-Gastaut prove seizure frequency in Lennox-Gastaut syndrome,
syndrome, the PMEs, and the infantile epileptic en- symptomatic generalized epilepsy, and Unverricht-
cephalopathies (45,100–102,138–144). Lundborg disease, although no prospective, controlled
Piracetam is effective and well tolerated as add-on ther- studies have investigated its use in any of these
apy for the treatment of myoclonus associated with the seizure/epilepsy types (65–69,111,152,157–159). Fur-
PMEs (107,145). The drug’s efficacy is reported to be thermore, such treatment primarily leads to improvement
highest within the first 12 months and subsequently stabi- in generalized convulsive seizures, rather than improve-
lizes (146–149). Treatment is usually initiated at a dosage ment in myoclonus.
of 3.2 g/day, divided in three doses and increased by Finally, the ketogenic diet has shown efficacy in the
2.4 g/day every week to a stable clinical response or a treatment of myoclonic seizures associated with differing
maximum dosage of 20 g/day (mean = 13.5 g/day), ex- etiologies and is the treatment of choice for glucose trans-
ceptionally up to 40 g/day. porter protein deficiency (46,64,65,160–162). The keto-
LEV is chemically similar to piracetam and is effective genic diet has also been useful in treating Lennox-Gastaut
in preventing the photoparoxysmal response in patients syndrome, for which its efficacy may be comparable to
with photosensitive epilepsy (150), which may predict its drug therapy (65,161). Additionally, open-label studies
efficacy in myoclonic seizures. Anecdotal reports suggest have demonstrated that the ketogenic diet can produce dra-
efficacy of LEV for myoclonus in postencephalitic and matic improvements for children with symptomatic my-
postanoxic myoclonus, Unverricht-Lundborg disease, and oclonic epilepsy and infantile spasms (46,162).
treatment-resistant JME (106,125–127,151).
CONCLUSIONS
100 Despite the availability of numerous treatment options,
≤ 50% 100% the diagnosis and treatment of myoclonic seizures con-
63%
67% tinue to be challenging. First, the clinician must determine
Patients (%)

if the events that a patient experiences are indeed my-


50 48%
oclonic seizures. Next, it is important to define the etiology
33%
of the episodes. For some rare conditions, special diets or
16%
10% vitamins may be used to treat the underlying etiology, and
0 no other therapies may be needed. However, most cases
GTC
(n=96)
Myoclonic
(n=21)
Absence
(n=50)
GTC
(n=96)
Myoclonic
(n=21)
Absence
(n=50)
require other treatments, and identification of the epilepsy
syndrome and its etiology is critical for making decisions
FIG. 5. Primary generalized epilepsy treated with topiramate:
seizure reduction for ≥6 months (61). GTC, generalized tonic- regarding therapy. Comorbidities must also be considered,
clonic. and treatment should be individualized for each patient.

Epilepsia, Vol. 44, Suppl. 11, 2003


34 J. W. WHELESS AND R. SANKAR

TABLE 1. Antiepileptic drug-related exacerbation of 11. Swinyard EA, Sofia RD, Kupferberg HJ. Comparative anticon-
myoclonic seizuresa vulsant activity and neurotoxicity of felbamate and four proto-
type antiepileptic drugs in mice and rats. Epilepsia 1986;27:27–
Drug Epilepsy syndrome 34.
12. Taylor CP. Gabapentin: mechanism of action. In: Levy RH, Matt-
Carbamazepine Angelman syndrome son RH, Meldrum BS, eds. Antiepileptic drugs. 4th ed. New York:
Autosomal dominant cortical Raven Press, 1995:829–41.
myoclonus and epilepsy 13. Holmes GL, Pearl PL. Gabapentin. In: Pellock JM, Dodson WE,
Juvenile myoclonic epilepsy Bourgeois BFD, eds. Pediatric epilepsy: diagnosis and ther-
Severe myoclonic epilepsy in infancy apy. 2nd ed. New York: Demos Medical Publishing, 2001:453–
Myoclonic-astatic epilepsy 9.
Wolf-Hirschhorn syndrome (4p-) 14. Sankar R, Derdiarian AT. Vigabatrin. CNS Drug Rev 1998;4:260–
Phenytoin Myoclonic-astatic epilepsy 74.
Progressive myoclonic epilepsies 15. Kanthasamy AG, Vu TQ, Yun RJ, Truong DD. Antimyoclonic ef-
Juvenile myoclonic epilepsy fect of gabapentin in a posthypoxic animal model of myoclonus.
Eur J Pharmacol 1996;297:219–24.
Lamotrigine Severe myoclonic epilepsy in infancy 16. Miller AA, Wheatley P, Sawyer DA, Baxter MG, Roth B. Phar-
Lennox-Gastaut syndrome macological studies on lamotrigine, a novel potential antiepilep-
Juvenile myoclonic epilepsy tic drug: 1. Anticonvulsant profile in mice and rats. Epilepsia
Vigabatrin Lennox-Gastaut syndrome 1986;27:483–9.
Myoclonic-astatic epilepsy 17. Buchanan N. The use of lamotrigine in juvenile myoclonic
Juvenile myoclonic epilepsy epilepsy. Seizure 1996;5:149–51.
Severe myoclonic epilepsy in infancy 18. Guerrini R, Dravet C, Genton C, Belmonte A, Kaminska A, Du-
Gabapentin Lennox-Gastaut syndrome lac O. Lamotrigine and seizure aggravation in severe myoclonic
Partial seizures epilepsy. Epilepsia 1998;39:508–12.
19. Guerrini R, Belmonte A, Parmeggiani L, Perucca E. Myoclonic sta-
Pregabalin Partial seizures tus epilepticus following high-dosage lamotrigine therapy. Brain
Generalized seizures Dev 1999;21:420–4.
a See 20. Janszky J, Rasonyi G, Halasz P, et al. Disabling erratic myoclonus
reference 18, 19, 34, 71, 72, 77, 91–94, and 163–166. during lamotrigine therapy with high serum level—report of two
cases. Clin Neuropharmacol 2000;23:86–9.
21. Shank RP, Gardocki JF, Vaught JL, et al. Topiramate: preclin-
ical evaluation of a structurally novel anticonvulsant. Epilepsia
Both pharmacologic and nonpharmacologic therapeutic 1994;35:450–60.
options are available. The neurologist must be aware that 22. Killiam KF, Killiam EK, Naquet R. Mise en evidence chez certains
singes d’un syndrome myoclonique [in French]. C R Acad Sci
many AEDs have been reported to cause exacerbation (Paris) 1966;262:1010–2.
of myoclonic seizures (Table 1) (18,19,34,71,72,77,91– 23. Meldrum B, Wilkins AJ. Photosensitive epilepsy in man and
94,163–166). Because myoclonic seizures can be difficult the baboon: integration of pharmacological and psychosocial ev-
idence. In: Schwartzkroin PA, Wheal HV, eds. Electrophysiology
to treat, physicians need to be flexible in their approach of epilepsy. London: Academic Press, 1984:51–77.
and tailor therapy to each patient. 24. McLean MJ. Gabapentin. In: Wiley E, ed. The treatment of
epilepsy: principles and practice. 2nd ed. Baltimore: Williams &
Wilkins, 1997:884–898.
REFERENCES 25. Meldrum BS, Horton R. Blockade of epileptic responses in the pho-
tosensitive baboon, Papio papio, by two irreversible inhibitors of
1. Jeavons PM, Clark JE, Maheshwari MC. Treatment of generalized GABA-transaminase, gamma-acetylenic GABA (4-amino-hex-5-
epilepsies of childhood and adolescence with sodium valproate ynoic acid) and gamma-vinyl GABA (4-amino-hex-5-enoic acid).
(“epilim”). Dev Med Child Neurol 1977;19:9–25. Psychopharmacology 1978;59:47–50.
2. Wallace SJ. Myoclonus and epilepsy in childhood: a review of treat- 26. Gundlach AL. Disorder of the inhibitory glycine receptor: inherited
ment with valproate, ethosuximide, lamotrigine and zonisamide. myoclonus in Poll Hereford calves. FASEB J 1990;4:2761–6.
Epilepsy Res 1998;29:147–54. 27. Avanzini G, Fariello R. Animal models of epilepsies. In: Avanzini
3. Dulac O. Epileptic encephalopathy. Epilepsia 2001;42(suppl G, Regesta G, Tanganelli P, Avoli M, eds. Molecular and cellular
3):23–6. targets for anti-epileptic drugs. London: John Libbey Eurotext,
4. Hallett M. Myoclonus and myoclonic syndromes. In: Engel J, 1997:91–109.
Pedley TA, eds. Epilepsy: a comprehensive textbook. Philadelphia: 28. Becker L, von Wegerer J, Schenkel J, Zeilhofer HU, Swandulla
Lippincott-Raven, 1997:2717–23. D, Weiher H. Disease-specific human glycine receptor alpha1
5. Janz D, Inoue Y, Seino M. Myoclonic seizures. In: Engel J, subunit causes hyperekplexia phenotype and impaired glycine-
Pedley TA, eds. Epilepsy: a comprehensive textbook. Philadelphia: and GABAA -receptor transmission in transgenic mice. J Neurosci
Lippincott-Raven, 1997:591–603. 2002;22:2505–12.
6. ILAE Commission report. Myoclonus and epilepsy in childhood. 29. AAN Practice Guidelines. American Academy of Neurology
Epilepsia 1997;38:1251–4. Web site. Available at: http//www.aan.com/professionals/practice/
7. Wallace SJ. Newer antiepileptic drugs: advantages and disadvan- index.cfm. (Accessed January 6, 2003.)
tages. Brain Dev 2001;23:277–83. 30. Ohtahara S, Yamatogi Y. Severe encephalopathic epilepsy in early
8. White HS, Woodhead JH, Franklin MR, et al. Experimental selec- infancy. In: Pellock J, Dodson WE, Bourgeois BFD, eds. Pedi-
tion, quantification, and evaluation of antiepileptic drugs. In: Levy atric epilepsy: diagnosis and therapy. 2nd ed. New York: Demos,
RH, Mattson RH, Meldrum BS, eds. Antiepileptic drugs. 4th ed. 2001:193–9.
New York: Raven Press, 1995:99–122. 31. Trevathan E. Infantile spasms and Lennox-Gastaut syndrome.
9. Ferrendelli JA, Holland KD, McKeon AC, Covey DF. Comparison J Child Neurol 2002;17(suppl 2):2S9–22.
of the anticonvulsant activities of ethosuximide, valproate, and a 32. Mackay M, Weiss S, Snead OC III. Treatment of infantile spasms:
new anticonvulsant, thiobutyrolactone. Epilepsia 1989;30:617–22. an evidence-based approach. Int Rev Neurobiol 2002;49:157–84.
10. Rho JM, Sankar R. The pharmacologic basis of antiepileptic drug 33. Dulac O, Chiron C, Luna D, et al. Vigabatrin in childhood epilepsy.
action. Epilepsia 1999;40:1471–83. J Child Neurol 1991;6(suppl):2S30–7.

Epilepsia, Vol. 44, Suppl. 11, 2003


MYOCLONUS TREATMENT STRATEGIES 35

34. Luna D, Dulac O, Pajot N, Beaumont D. Vigabatrin in the treatment 62. Sachdeo RC, Glauser TA, Ritter F, Reife R, Lim P, Pledger
of childhood epilepsies: a single-blind placebo-controlled study. G. A double-blind, randomized trial of topiramate in Lennox-
Epilepsia 1989;30:430–7. Gastaut syndrome. Topiramate YL Study Group. Neurology
35. Elterman RD, Shields WD, Mansfield KA, Nakagawa J. Random- 1999;52:1882–7.
ized trial of vigabatrin in patients with infantile spasms. Neurology 63. Coppola G, Caliendo G, Veggiotti P, et al. Topiramate as add-
2001;57:1416–21. on drug in children, adolescents and young adults with Lennox-
36. Curatolo P, Seri S, Verdecchia M, Bombardieri R. Infantile spasms Gastaut syndrome: an Italian multicentric study. Epilepsy Res
in tuberous sclerosis complex. Brain Dev 2001;23:502–7. 2002;51:147–53.
37. Curatolo P, Verdecchia M, Bombardieri R. Vigabatrin for tuberous 64. Vining EP. Clinical efficacy of the ketogenic diet. Epilepsy Res
sclerosis complex. Brain Dev 2001;23:649–53. 1999;37:181–90.
38. Hancock E, Osborne JP, Milner P. The treatment of West syn- 65. Wheless JW, Baumgartner J, Ghanbari C. Vagus nerve stimulation
drome: a Cochrane review of the literature to December 2000. and the ketogenic diet. Neurol Clin 2001;19:371–407.
Brain Dev 2001;23:624–34. 66. Parker AP, Polkey CE, Binnie CD, Madigan C, Ferrie CD, Robin-
39. Mikati MA, Lepejian GA, Holmes GL. Medical treatment of pa- son RO. Vagal nerve stimulation in epileptic encephalopathies. Pe-
tients with infantile spasms. Clin Neuropharmacol 2002;25:61–70. diatrics 1999;103:778–82.
40. Dreifuss F, Farwell J, Holmes G, et al. Infantile spasms. Com- 67. Frost M, Gates J, Helmers SL, et al. Vagus nerve stimulation in
parative trial of nitrazepam and corticotropin. Arch Neurol children with refractory seizures associated with Lennox-Gastaut
1986;43:1107–10. syndrome. Epilepsia 2001;42:1148–52.
41. Toribe Y. High-dose vitamin B6 treatment in West syndrome. Brain 68. Wheless JW. Vagus nerve stimulation. In: Wyllie E, ed. The
Dev 2001;23:654–57. treatment of epilepsy: principles and practice. 3rd ed. Baltimore:
42. Gospe SM. Pyridoxine-dependent seizures: findings from recent Lippincott, Williams and Wilkins, 2001:1007–15.
studies pose new questions. Pediatr Neurol 2002;26:181–5. 69. Majoie HJ, Berfelo MW, Aldenkamp AP, Evers SM, Kessels AG,
43. Baxter P. Pyridoxine-dependent and pyridoxine-responsive Renier WO. Vagus nerve stimulation in children with therapy-
seizures. Dev Med Child Neurol 2001;43:416–20. resistant epilepsy diagnosed as Lennox-Gastaut syndrome: clinical
44. Nabbout R. A risk-benefit assessment of treatments for infantile results, neuropsychological effects, and cost-effectiveness. J Clin
spasms. Drug Saf 2001;24:813–28. Neurophysiol 2001;18:419–28.
45. Suzuki Y, Imai K, Toribe Y, et al. Long-term response to zon- 70. Helmers SL, Wheless JW, Frost M, et al. Vagus nerve stimulation
isamide in patients with West syndrome. Neurology 2002;58:1556– therapy in pediatric patients with refractory epilepsy: retrospective
9. study. J Child Neurol 2001;16:843–8.
46. Kossoff EH, Pyzik PL, McGrogan JR, Vining EP, Freeman JM. 71. Vossler DG. Exacerbation of seizures in Lennox-Gastaut syndrome
Efficacy of the ketogenic diet for infantile spasms. Pediatrics by gabapentin. Neurology 1996;46:852–3.
2002;109:780–3. 72. Lortie A, Chiron C, Mumford J, Dulac O. The potential for
47. Chiron C, Marchand MC, Tran A, et al. Stiripentol in se- increasing seizure frequency, relapse, and appearance of new
vere myoclonic epilepsy in infancy: a randomized placebo- seizure types with vigabatrin. Neurology 1993;43(suppl 5):S24–
controlled syndrome-dedicated trial. STICLO study group. Lancet 7.
2000;356:1638–42. 73. Bonanni P, Parmeggiani L, Guerrini R. Different neurophysiologic
48. Perez J, Chiron C, Musial C, et al. Stiripentol: efficacy and tolera- patterns of myoclonus characterize Lennox-Gastaut syndrome and
bility in children with epilepsy. Epilepsia 1999;40:1618–26. myoclonic astatic epilepsy. Epilepsia 2002;43:609–15.
49. Todt H, Muller D. The therapy of benign myoclonic epilepsy in 74. Arzimanoglou A. Treatment options in pediatric epilepsy syn-
infants. Epilepsy Res Suppl 1992;6:137–9. dromes. Epileptic Disord 2002;4:217–25.
50. Clancy RR. Valproate: an update—the challenge of modern pedi- 75. Oguni H, Tanaka T, Hayashi K, et al. Treatment and long-term
atric seizure management. Curr Prob Pediatr 1990;20:161–233. prognosis of myoclonic-astatic epilepsy of early childhood. Neu-
51. Wheless JW, Constantinou JE. Lennox-Gastaut syndrome. Pediatr ropediatrics 2002;33:122–32.
Neurol 1997;17:203–11. 76. Dulac O, Kaminska A. Use of lamotrigine in Lennox-Gastaut and
52. Jeavons PM, Clark JE. Sodium valproate in treatment of epilepsy. related epilepsy syndromes. J Child Neurol 1997;12(suppl 1):S23–
Br Med J 1974;2:584–6. 8.
53. Covanis A, Gupta AK, Jeavons PM. Sodium valproate monother- 77. Perucca E, Gram L, Avanzini G, Dulac O. Antiepileptic drugs as a
apy and polytherapy. Epilepsia 1982;23:693–720. cause of worsening seizures. Epilepsia 1998;39:5–17.
54. Henriksen O, Johannessen SI. Clinical and pharmacokinetic ob- 78. Buoni S, Grosso S, Fois A. Lamotrigine in typical absence epilepsy.
servations on sodium valproate: a 5-year follow-up study in 100 Brain Dev 1999;21:303–6.
children with epilepsy. Acta Neurol Scand 1982;65:504–23. 79. Beran RG, Berkovic SF, Dunagan FM, et al. Double-blind,
55. The Felbamate Study Group in Lennox-Gastaut Syndrome. Effi- placebo-controlled, crossover study of lamotrigine in treatment-
cacy of felbamate in childhood epileptic encephalopathy (Lennox- resistant generalised epilepsy. Epilepsia 1998;39:1329–33.
Gastaut syndrome). N Engl J Med 1993;328:29–33. 80. Wolf P. Juvenile myoclonic epilepsy. In: Roger J, Dravet C, Bureau
56. Siegel H, Kelley K, Stertz B, et al. The efficacy of felbamate as add- M, Dreifuss FE, Wolf P, eds. Epileptic syndromes in infancy, child-
on therapy to valproic acid in Lennox-Gastaut syndrome. Epilepsy hood and adolescence. London: John Libbey Eurotext, 1992:313–
Res 1999;34:91–7. 27.
57. Motte J, Trevathan E, Arvidsson JF, Barrera MN, Mullens EL, 81. Messenheimer J. Efficacy and safety of lamotrigine in pediatric
Manasco P. Lamotrigine for generalized seizures associated with patients. J Child Neurol 2002;17(suppl 2):2S34–42.
the Lennox-Gastaut syndrome. Lamictal Lennox-Gastaut Study 82. Cross JH. Topiramate monotherapy for childhood absence seizures:
Group [published correction appears in N Engl J Med. 1998; an open label pilot study. Seizure 2002;11:406–10.
339:851–2]. N Engl J Med 1997;337:1807–12. 83. Tassinari CA, Bureau M, Thomas P. Epilepsy with myoclonic ab-
58. Donaldson JA, Glauser TA, Olberding LS. Lamotrigine adjunctive sences. In: Roger J, Bureau M, Dravet C, Dreifuss FE, Perret A,
therapy in childhood epileptic encephalopathy (the Lennox Gastaut Wolf P, eds. Epileptic syndromes in infancy, childhood, and ado-
syndrome). Epilepsia 1997;38:68–73. lescence. London: John Libbey Eurotext, 1992:151–60.
59. Glauser TA, Levisohn PM, Ritter F, Sachdeo RC. Topiramate in 84. Richens A. Treatment of eyelid myoclonia with absences. In:
Lennox-Gastaut syndrome: open-label treatment of patients com- Duncan JS, Panayiotopoulos CP, eds. Eyelid myoclonia with ab-
pleting a randomized controlled trial. Topiramate YL Study Group. sences. London: John Libbey Eurotext, 1996:117–20.
Epilepsia 2000;41(suppl 1):S86–90. 85. Delgado-Escueta AV, Enrile-Bacsal F. Juvenile myoclonic
60. Glauser TA. Topiramate in the catastrophic epilepsies of childhood. epilepsy of Janz. Neurology 1984;34:285–94.
J Child Neurol 2000;15:S14–21. 86. Penry JK, Dean JC, Riela AR. Juvenile myoclonic epilepsy:
61. Wheless JW. Use of topiramate in childhood generalized seizure long-term response to therapy. Epilepsia 1989;30(suppl 4):S19–
disorders. J Child Neurol 2000;15(suppl 1):S7–13. 23.

Epilepsia, Vol. 44, Suppl. 11, 2003


36 J. W. WHELESS AND R. SANKAR

87. Bourgeois B, Beaumanoir A, Blajev B, et al. Monotherapy with val- 112. Berkovic SF, Cochius J, Andermann E, Andermann F. Progres-
proate in primary generalized epilepsies. Epilepsia 1987;28(suppl sive myoclonus epilepsies: clinical and genetic aspects. Epilepsia
2):S8–11. 1993;34(suppl 3):S19–30.
88. Grunewald RA, Panayiotopoulos CP. Juvenile myoclonic epilepsy. 113. Scheffer IE, Berkovic SF. Generalized epilepsy with febrile
A review. Arch Neurol 1993;50:594–8. seizures plus. A genetic disorder with heterogeneous clinical phe-
89. Biton V, Montouris GD, Ritter F, et al. A randomized placebo- notypes. Brain 1997;120:479–90.
controlled study of topiramate in primary generalized tonic-clonic 114. Singh R, Scheffer IE, Crossland K, Berkovic SF. Generalized
seizures. Topiramate YTC Study Group. Neurology 1999;52:1330– epilepsy with febrile seizures plus: a common childhood-onset ge-
7. netic epilepsy syndrome. Ann Neurol 1999;45:75–81.
90. Resor SR Jr, Resor LD. Chronic acetazolamide monotherapy 115. Pueschel SM, Louis S, McKnight P. Seizure disorders in Down
in the treatment of juvenile myoclonic epilepsy. Neurology syndrome. Arch Neurol 1991;48:318–20.
1990;40:1677–81. 116. Stafstrom CE, Patxot OF, Gilmore HE, Wisniewski KE. Seizures
91. Genton P, Gelisse P, Thomas P, Dravet C. Do carbamazepine in children with Down syndrome: etiology, characteristics and out-
and phenytoin aggravate juvenile myoclonic epilepsy? Neurology come. Dev Med Child Neurol 1991;33:191–200.
2000;55:1106–9. 117. Silva ML, Cieuta C, Guerrin R, Plouin P, Livet MO, Dulac O. Early
92. Biraben A, Allain H, Scarabin JM, Schuck S, Edan G. Exacerba- clinical and EEG features of infantile spasms in Down syndrome.
tion of juvenile myoclonic epilepsy with lamotrigine. Neurology Epilepsia 1996;37:977–82.
2000;55:1757–8. 118. Goldberg-Stern H, Strawsburg RH, Patterson B, et al. Seizure fre-
93. Carrazana EJ, Wheeler SD. Exacerbation of juvenile myoclonic quency and characteristics in children with Down syndrome. Brain
epilepsy with lamotrigine. Neurology 2001;56:1424–5. Dev 2001;23:375–8.
94. Guerrini R, Bonanni P, Patrignani A, et al. Autosomal dominant 119. Moller JC, Hamer HM, Oertel WH, Rosenow F. Late-onset
cortical myoclonus and epilepsy (ADCME) with complex partial myoclonic epilepsy in Down’s syndrome (LOMEDS). Seizure
and generalized seizures. A newly recognized epilepsy syndrome 2002;11(suppl A):303–5.
with linkage to chromosome 2p11.1–q12.2. Brain 2001;124:2459– 120. Battaglia A, Carey JC, Wright TJ. Wolf-Hirschhorn (4p-) syn-
75. drome. Adv Pediatr 2001;48:75–113.
95. Plaster NM, Uyama E, Uchino M, et al. Genetic localization of the 121. Guerrini R, Gobbi G, Genton P, et al. Chromosomal abnormalities.
familial adult myoclonic epilepsy (FAME) gene to chromosome In: Engel J, Pedley TA, eds. Epilepsy: a comprehensive textbook.
8q24. Neurology 1999;53:1180–3. Philadelphia: Lippincott-Raven, 1997:2533–46.
96. Mikami M, Yasuda T, Terao A, et al. Localization of a gene for 122. Franz DN, Glauser TA, Todor C, Williams S. Topiramate ther-
benign adult familial myoclonic epilepsy to chromosome 8q23.3- apy of epilepsy associated with Angelman’s syndrome. Neurology
q24.1. Am J Hum Genet 1999;65:745–51. 2000;54:1185–8.
97. Labauge P, Amer LO, Simonetta-Moreau M, et al. Absence of 123. Boles RG, Seashore MR, Mitchell WG, Kollros PR, Mofidi
linkage to 8q24 in a European family with familial adult myoclonic S, Novotny EJ. Glucose transporter type 1 deficiency: a study
epilepsy (FAME). Neurology 2002;58:941–4. of two cases with video-EEG. Eur J Pediatr 1999;158:978–
98. Conry JA. Progressive myoclonic epilepsies. J Child Neurol 83.
2002;17(suppl 1):S80–4. 124. De Vivo DC, Trifiletti RR, Jacobson RI, Ronen GM, Behmand
99. Wheless JW, Kim HL. Adolescent seizures and epilepsy syn- RA, Harik SI. Defective glucose transport across the blood-
dromes. Epilepsia 2002;43(suppl 3):33–52. brain barrier as a cause of persistent hypoglycorrhachia,
100. Yoshimura I, Kaneko S, Yoshimura N, Murakami T. Long-term seizures, and developmental delay. N Engl J Med 1991;325:703–
observations of two siblings with Lafora disease treated with zon- 9.
isamide. Epilepsy Res 2001;46:283–7. 125. Krauss GL, Bergin A, Kramer RE, Cho YW, Reich SG. Suppres-
101. Henry TR, Leppik IE, Gumnit RJ, Jacobs M. Progres- sion of post-hypoxic and post-encephalitic myoclonus with leve-
sive myoclonus epilepsy treated with zonisamide. Neurology tiracetam. Neurology 2001;56:411–2.
1988;38:928–31. 126. Frucht SJ, Louis ED, Chuang C, Fahn S. A pilot tolerability and
102. Kyllerman M, Ben-Menachem E. Zonisamide for progressive efficacy study of levetiracetam in patients with chronic myoclonus.
myoclonus epilepsy: long-term observations in seven patients. Neurology 2001;57:1112–4.
Epilepsy Res 1998;29:109–14. 127. Schauer R, Singer M, Saltuari L, Kofler M. Suppression of cortical
103. Hirano M, DiMauro S. Primary mitochondrial disease. In: Engel J, myoclonus by levetiracetam. Mov Disord 2002;17:411–5.
Pedley TA, eds. Epilepsy: a comprehensive textbook. Philadelphia: 128. Coulter DL, Wu H, Allen RJ. Valproic acid therapy in childhood
Lippincott-Raven, 1997:2563–70. epilepsy. JAMA 1980;244:785–8.
104. Aberg L, Kirveskari E, Santavuori P. Lamotrigine therapy in juve- 129. Sotaniemi K. Valproic acid in the treatment of nonepileptic my-
nile neuronal ceroid lipofuscinosis. Epilepsia 1999;40:796–9. oclonus. Arch Neurol 1982;39:448–9.
105. Aberg L, Heiskala H, Vanhanen SL, et al. Lamotrigine therapy in 130. Sheth RD, Gidal BE. Intravenous valproic acid for myoclonic status
infantile neuronal ceroid lipofuscinosis (INCL). Neuropediatrics epilepticus. Neurology 2000;54:1201.
1997;28:77–9. 131. Gericke CA, Picard F, Saint-Martin A, Strumia S, Marescaux C,
106. Genton P, Gelisse P. Antimyoclonic effect of levetiracetam. Epilep- Hirsch E. Efficacy of lamotrigine in idiopathic generalized epilepsy
tic Disord 2000;2:209–12. syndromes: a video-EEG-controlled, open study. Epileptic Disord
107. Fedi M, Reutens D, Dubeau F, Andermann E, D’Agostino D, 1999;1:159–65.
Andermann F. Long-term efficacy and safety of piracetam in 132. Besag FM, Wallace SJ, Dulac O, Alving J, Spencer SC, Hosking G.
the treatment of progressive myoclonus epilepsy. Arch Neurol Lamotrigine for the treatment of epilepsy in childhood. J Pediatr
2001;58:781–6. 1995;127:991–7.
108. Genton P, Guerrini R. Antimyoclonic effects of alcohol in progres- 133. Schlumberger E, Chavez F, Palacios L, Rey E, Pajot N, Dulac O.
sive myoclonus epilepsy. Neurology 1990;40:1412–6. Lamotrigine in treatment of 120 children with epilepsy. Epilepsia
109. Hurd RW, Wilder BJ, Helveston WR, Uthman BM. Treat- 1994;35:359–67.
ment of four siblings with progressive myoclonus epilepsy of 134. Veggiotti P, Cieuta C, Rex E, Dulac O. Lamotrigine in infantile
the Unverricht-Lundborg type with N-acetylcysteine. Neurology spasms. Lancet 1994;344:1375–6.
1996;47:1264–8. 135. Pisani F, Oteri G, Russo MF, Di Perri R, Perucca E, Richens A. The
110. Edwards MJ, Hargreaves IP, Heales SJ, et al. N-acetylcysteine and efficacy of valproate-lamotrigine comedication in refractory com-
Unverricht-Lundborg disease: variable response and possible side plex partial seizures: evidence for a pharmacodynamic interaction.
effects. Neurology 2002;59:1447–9. Epilepsia 1999;40:1141–6.
111. Smith B, Shatz R, Elisevich K, Bespalova IN, Burmeister M. Ef- 136. Nieto-Barrera M, Candau R, Nieto-Jimenez M, Correa A, del Portal
fects of vagus nerve stimulation on progressive myoclonus epilepsy LR. Topiramate in the treatment of severe myoclonic epilepsy in
of Unverricht-Lundborg type. Epilepsia 2000;41:1046–8. infancy. Seizure 2000;9:590–4.

Epilepsia, Vol. 44, Suppl. 11, 2003


MYOCLONUS TREATMENT STRATEGIES 37

137. Uldall P, Buchholt JM. Clinical experiences with topiramate tion in six patients with hypothalamic hamartomas. Pediatr Neurol
in children with intractable epilepsy. Eur J Paediatr Neurol 2000;23:167–8.
1999;3:105–11. 153. Maggio V, Wheless J. Dietary treatment of seizures from a hy-
138. Yagi K, Seino M. Methodological requirements for clinical tri- pothalamic hamartoma. In: Schmidt D, Schachter SC, eds. 110
als in refractory epilepsies–our experience with zonisamide. Prog Puzzling cases of epilepsy. London: Martin Dunitz, 2001:434–8.
Neuropsychopharmacol Biol Psychiatry 1992;16:79–85. 154. Regis J, Bartolomei F, de Toffol B, et al. Gamma knife surgery
139. Leppik IE. Zonisamide. Epilepsia 1999;40(suppl 5):S23–9. for epilepsy related to hypothalamic hamartomas. Neurosurgery
140. Yanai S, Hanai T, Narazaki O. Treatment of infantile spasms with 2000;47:1343–51.
zonisamide. Brain Dev 1999;21:157–61. 155. Kramer U, Spector S, Nasser W, Siomin V, Fried I, Constantini
141. Kishi T, Nejihashi Y, Kajiyama M, Ueda K. Successful zon- S. Surgical treatment of hypothalamic hamartoma and refractory
isamide treatment for infants with hypsarrhythmia. Pediatr Neurol seizures: a case report and review of the literature. Pediatr Neuro-
2000;23:274–7. surg 2001;34:40–2.
142. Suzuki Y, Nagai T, Ono J, et al. Zonisamide monotherapy in newly 156. Palmini A, Chandler C, Andermann F, et al. Resection of the le-
diagnosed infantile spasms. Epilepsia 1997;38:1035–8. sion in patients with hypothalamic hamartomas and catastrophic
143. Sakamoto K, Kurokawa T, Tomita S, et al. Effects of zonisamide epilepsy. Neurology 2002;58:1338–47.
in children with epilepsy. Curr Therapeutic Res 1988;13:378–83. 157. Labar D. Vagus nerve stimulation for intractable epilepsy in chil-
144. Glauser TA, Pellock JM. Zonisamide in pediatric epilepsy: review dren. Dev Med Child Neurol 2000;42:496–9.
of the Japanese experience. J Child Neurol 2002;17:87–96. 158. Labar D, Nikolov B, Tarver B, Fraser R. Vagus nerve stimula-
145. Genton P, Van Vleymen B. Piracetam and levetiracetam: close tion for symptomatic generalized epilepsy: a pilot study. Epilepsia
structural similarities but different pharmacological and clinical 1998;39:201–5.
profiles. Epileptic Disord 2000;2:99–105. 159. Wheless JW, Maggio V. Vagus nerve stimulation therapy in pa-
146. Uthman BM, Reichl A. Progressive myoclonic epilepsies. Curr tients younger than 18 years. Neurology 2002;59(suppl 4):S21–5.
Treat Options Neurol 2002;4:3–17. 160. Katyal NG, Koehler AN, McGhee B, Foley CM, Crumrine PK. The
147. Obeso JA, Artieda J, Quinn N, et al. Piracetam in the treatment of ketogenic diet in refractory epilepsy: the experience of Children’s
different types of myoclonus. Clin Neuropharmacol 1988;11:529– Hospital of Pittsburgh. Clin Pediatr (Phila) 2000;39:153–9.
36. 161. Freeman JM, Vining EP, Pillas DJ, Pyzik PL, Casey JC, Kelly LM.
148. Brown P, Steiger MJ, Thompson PD, et al. Effectiveness of pirac- The efficacy of the ketogenic diet—1998: a prospective evaluation
etam in cortical myoclonus. Mov Disord 1993;8:63–8. of intervention in 150 children. Pediatrics 1998;102:1358–63.
149. Genton P, Guerrini R, Remy P. Piracetam in the treatment of 162. Nordli DR Jr, Kuroda MM, Carroll J, et al. Experience with the
cortical myoclonus. Pharmacopsychiatry 1999;32(suppl 1):49– ketogenic diet in infants. Pediatrics 2001;108:129–33.
53. 163. Huppertz HJ, Feuerstein TJ, Schulze-Bonhage A. Myoclonus in
150. Kastelijn-Nolst Trenite DG, Marescaux C, Stodieck S, Edelbroek epilepsy patients with anticonvulsive add-on therapy with prega-
PM, Oosting J. Photosensitive epilepsy: a model to study the ef- balin. Epilepsia 2001;42:790–2.
fects of antiepileptic drugs. Evaluation of the piracetam analogue, 164. Scheyer RD, Assaf BA, Spencer SS, Mattson RH. Gabapentin re-
levetiracetam. Epilepsy Res 1996;25:225–30. lated myoclonus [Abstract]. Epilepsia 1996;37(suppl 5):203.
151. Smith K, Betts T, Greenhill L. Levetiracetam: a promising option 165. Viani F, Romeo A, Viri M, et al. Seizure and EEG patterns in
for the treatment of juvenile myoclonic epilepsy. Presented at: 4th Angelman’s syndrome. J Child Neurol 1995;10:467–71.
European Congress on Epileptology; October 11, 2000; Firenze. 166. Asconape J, Diedrich A, DellaBadia J. Myoclonus associated with
152. Murphy JV, Wheless JW, Schmoll CM. Left vagal nerve stimula- the use of gabapentin. Epilepsia 2000;41:479–81.

Epilepsia, Vol. 44, Suppl. 11, 2003

Você também pode gostar