Sunteți pe pagina 1din 6

Reviews

Vagus-nerve stimulation for the treatment of


epilepsy

Elinor Ben-Menachem

Vagus-nerve stimulation (VNS) is now an accepted treatment stimulators and were given intermittent stimulation of a 30 s
for patients with refractory epilepsy. There have been many train of impulses (on) every 5 min (off). The positive results of
studies suggesting that VNS affects the brain in such areas these pilot studies led to double-blind, low-dose, placebo-
as the thalamus and other limbic structures. In addition, controlled trials to elucidate further the efficacy of VNS in
there is some evidence that norepinephrine is important in partial seizures.17,18
the prophylactic antiseizure effects of VNS. The efficacy of
VNS has been established for partial seizure types, even in Anatomy
refractory patients who did not respond to surgical treatment Studies in cats have shown that the vagus nerve consists of
for epilepsy. There are also data, from open-label studies, 80% afferent fibres projecting from the viscera to the nucleus
that suggest efficacy in other seizure types. Therefore, VNS tractus solitarius.19 Efferent fibres provide parasympathetic
seems to be a broad-spectrum treatment for epilepsy. innervation primarily to the lungs, heart, and gastrointestinal
Improvement is not immediate but increases over 18–24 tract, and they also innervate the voluntary muscles in the
months of treatment. Most studies report subjective larynx and pharynx. These fibres originate from the
improvements in various quality-of-life measurements during paraganglionic neurons in the dorsal motor nucleus of the
treatment with VNS—objective trials have confirmed this vagus nerve. There is an asymmetrical innervation to the
observation. Side-effects are mainly stimulation related and heart—the right vagus nerve innervates the sinoatrial node
reversible and they tend to decrease over time. They are and the left innervates the atrioventricular node. In dogs,
generally mild to moderate and seldom necessitate the when the right vagus nerve is stimulated, bradycardia is
removal of the device. No idiosyncratic side-effects have elicited to a greater degree than when the left nerve is
been reported in 12 years of experience, and VNS does not stimulated.20 For this reason, VNS implants are placed on the
interact with antiepileptic drugs. Most adverse events are left side of the neck and not the right side. Afferent fibres arise
predictable and related to the specific stimulation regimen. from receptors in the lungs, heart, aorta, gastrointestinal tract,
VNS does not have cognitive and systemic side-effects and and aortic chemoreceptors, and a small group arise from the
can, therefore, be a valuable treatment approach even for concha of the ear. The afferent fibres have their cell bodies in
patients who have poor tolerance of antiepileptic drugs. the nodose and jugular ganglia and project to the nucleus
tractus solitarius as well as to the dorsal motor nucleus, area
Lancet Neurology 2002; 1: 477–82 postrema, and nucleus cunneatus. From the nucleus tractus
solitarius there are synaptic connections to higher centres in
Corning created the first, although crude and external, vagus- the brain such as the hypothalamus, dorsal raphe, nucleus
nerve stimulator (VNS) in the 1880s. He lowered the number of ambigus, dorsal motor nucleus of the vagus nerve, amygdala,
seizures in patients treated with cardioinhibitory vagal and thalamus, which in turn project to the insular cortex.21
stimulation to reduce cerebral blood flow.1 VNS was, however, Positron-emission tomography and functional magnetic
forgotten as a useful antiseizure therapy until Zabara2–4 began to resonance imaging of the effects of VNS in human beings have
analyse the effect of stimulation in chemically induced seizures confirmed the influence the vagus nerve on higher brain
in dogs. The results were striking, and the technique has become structures. These studies have shown increases and decreases
an accepted and registered form of treatment for epilepsy; in blood flow in response to VNS. In one of the first positron-
worldwide, more than 15 000 patients receive this therapy. emission tomography studies, Ko and co-workers22 found that
That peripheral stimulation of the vagus nerve can affect VNS increased blood flow to the right thalamus, the right
the brain and cause striking changes in electroen- posterior temporal cortex, the left putamen, and the left
cephalographic patterns has been described several times since inferior cerebellum. In positron-emission tomography studies
the 1930s.5–10 Most animal experiments were done in cats that by Henry and colleagues23,24 blood flow was increased to the
had been given strychnine; VNS lowered the frequency of
spiking activity in this model. VNS was later shown to inhibit
seizures in the maximum electroshock and pentylenetetrazol
EBM is at the Department of Clinical Neuroscience, Sahlgrenska
models in rats11 and monkeys.12 A recent study suggests that Academy, Göteborg University, Sweden.
VNS also inhibits spiking activity in human encephalograms.13 Correspondence: Prof Elinor Ben-Menachem, Department of
Results of the first trial of VNS in human beings were Clinical Neuroscience, Sahlgrenska Academy, Göteborg University,
published in 199014,15 and 1993.16 In this trial, 16 patients with 413 45 Göteborg, Sweden. Tel +46 31 3421000;
refractory partial-onset seizures received vagus-nerve fax +46 31 211552; email ebm@neuro.gu.se

THE LANCET Neurology Vol 1 December 2002 http://neurology.thelancet.com 477

For personal use. Only reproduce with permission from The Lancet Publishing Group.
Review Vagus-nerve stimulation

Caudal Cephalad Mechanism of action


The mechanism of action of VNS is not fully understood.
Stimulation causes increases in cerebral blood flow and can
alter electroencephalographic patterns. CSF studies in
Anchor
human beings have shown changes in the concentrations of
tether several amino acids and neurotransmitters that might play
a part—especially ethanolamine, which is a sign of
increased turnover of membrane components.26 Perhaps
this is an indication of an increased amino-acid turnover in
a more general sense as is also implied by the observation of
high neuronal fos expression in the vagal areas of the
Negative
medulla (the locus coeruleus, the thalamic and
electrode hypothalamic nuclei, the amygdala, the cingulate cortex,
Positive and the retrosplenial cortex) in rats undergoing VNS.27
electrode There is fierce debate about whether conditions used in
human beings stimulate only A and B myelinated fibres or
whether C fibres, which are thin and unmyelinated, are also
needed for the antiepileptic effect. Evidence against the
influence of C fibres includes the lack of significant effect
on heart rate or blood pressure, and that there is no change
noted in gastrin concentrations or any other autonomic
function that would be expected if C fibres were stimulated
in human beings.18,28–30 In a recent study by Krahl and
colleagues,31 C fibres in rat vagal nerves were destroyed with
capsaicin. Despite the lack of C fibres, VNS had the same
striking efficacy in preventing pentelyntetrazol-induced
seizures.
Besides the increased fos expression in the locus
coeruleus, there is other evidence that this area is involved
in VNS antiseizure activity. When norepinephrine was
depleted by 6-hydroxydopamine infusion into the locus
coeruleus of rats, the seizure-suppressive effect of VNS was
abolished.32
In summary, current information suggests that VNS
Figure 1. Top: surgical implantation of VNS device. Bottom: position of activates neuronal networks in the thalamus and other limbic
VNS device once implanted.
structures and that norepinephrine may mediate the
inferior cerebellum, hypothalamus, and thalamus and antiseizure activity of VNS.
decreased in the areas of the hippocampus, amygdala, and
posterior cingulated gyrus during VNS. Generally, changes in The device and its implantation
blood flow were greater in the right hemisphere than in the The stimulation device is similar to a cardiac pacemaker and is
left. The main conclusions from these studies are that the implanted under the left clavicle in a procedure similar to that
thalamus is consistently involved, and that there are used to implant a pacemaker in the chest wall or more
pronounced changes in blood flow in the brain during VNS laterally in the axilla. Two helical bipolar stimulating
that correspond to anatomical synaptic projections. In recent electrodes33 are placed around the left vagus nerve after it has
experiments with functional magnetic resonance imaging, been exposed in the neck distal to the branching of the
Bohning and colleagues25 found that the areas of significant recurrent laryngeal nerve (figure 1). The left vagus nerve,
activation in response to VNS were the bilateral orbitofrontal never the right, is used to limit the risk of bradycardia or
and parieto-occipital cortex, the left temporal cortex, and the arrhythmias.20 The operation can be done under local or
left amygdala. Besides these specific areas there was a general general anaesthesia and takes about 1–2 h. Recovery is rapid,
diffuse increase in brain activity. and some centres do implantation on an outpatient basis.
The vagus nerve in the neck is enclosed in the carotid After the operation, the VNS can be turned on by use of a
sheath, which lies beneath the sternocleidomastoid muscle. In computer and program wand. The starting level of
the midcervical area the superior laryngeal nerve branches off stimulation is 0·25 mA in most cases; stimulation is increased
rostrally to the carotid bifurcation. This nerve is secondarily to 1·25–2·00 mA over several weeks. The most common
stimulated during VNS and can give rise to a feeling of tightness settings for the stimulator are frequency of 20–30 Hz (animal
or even pain in the throat area. The recurrent laryngeal nerve studies showed the best effect in the range of 10–60 Hz),4,12
travels alongside the vagal nerve, and hence is also affected by pulse width of 250–500 ␮s, time on of 30 s, and time off of
VNS causing vibration of the left vocal cord during stimulation 3–5 min. The times on and off can be changed, and many
and subsequent hoarseness during on times. clinicians use rapid cycling of 7 s on and 0·2 s off, thereby

478 THE LANCET Neurology Vol 1 December 2002 http://neurology.thelancet.com

For personal use. Only reproduce with permission from The Lancet Publishing Group.
Vagus-nerve stimulation
Review

increasing the amount of stimulation time given daily.


However, there is no hard evidence that one time setting is 80
better than the others. Increases in the amount of stimulation
70

Patients continuing (%)


will decrease battery life; therefore, high stimulation should be
used only if shown to be effective. Even the optimum current 60
can vary among individuals. Thus, after 12 years of use the
50
optimum VNS settings are still unknown.
40
Efficacy in epilepsy 30
Randomised, placebo-controlled, double-blind
20
studies
Two pivotal studies have attempted to answer the question 10
of whether VNS really does have an antiseizure effect. These 0
two trials also served as the main evidence for approval of VNS LVT TPM LTG
the US and European regulatory boards for the use of VNS Therapy
in patients with refractory partial-onset seizures.
The first trial (EO3)17 was an international multicentre
Figure 2. Comparison of long-term, 3-year continuation rates of patients
trial that included 114 patients over the age of 12 years who continuing treatment with VNS,34 levetiracetam (LVT),35 topiramate (TPM),
completed the prospective baseline phase of the study. All and lamotrigine (LTG).36
had more than six partial-onset seizures per month despite
treatment with antiepileptic drugs. 113 had devices and weaknesses associated with each analysis, the interesting
implanted and were randomly assigned to two groups. The finding was that the outcomes for VNS were similar for each
high-stimulation group received 30 s of VNS every 5 min analysis. In the last-visit carried forward analysis, by
(30 Hz, 500 ␮s, up to 3·5 mA) and the other group received 3 months after the end of the double-blind trial, patients had
30 s of VNS every 90 min (1 Hz, 130 ␮s, ⭐3·5 mA). No a median seizure reduction of 34%, and by the end of the
changes in the antiepileptic-drug treatments were allowed. year the median seizure reduction was 45% (p<0·0001). 34%
After 3 months of treatment with VNS, there was a 24·5% of patients had a seizure reduction of more than 50% and
decrease in seizures in the high-stimulation group compared 20% had a reduction of more than 75%. The stimulation
with 6·1% in the low-stimulation group (p=0·01). 31% of current was around 1·7 mA at the end of the year.
the high-stimulation group had seizure reduction of more In the EO1–EO5 study, 440 patients were followed up for
than 50% compared with 13% in the low-stimulation group. as long as 3 years. The same three analyses were used. The
In EO5,18 a multicentre USA based trial, 199 patients amount by which seizures were reduced increased from
with refractory partial seizures received implants and were baseline to 3 months into the follow-up phase and then at
followed up for 3 months. 103 patients were in the low- 2 years, after which it reached a plateau. Seizure reduction of
stimulation group and 95 were in the high-stimulation more than 50% was seen in 23% of patients at 3 months,
group with similar stimulation regimens to the EO3 study. 37% at 1 year, and 43% at both 2 years and 3 years.
After 3 months of stimulation there was a 28% decrease in Continuation rates are shown in figure 2, referenced in
seizures in the high-stimulation group and a 15% decrease in comparison with published information on the newer
the low-stimulation group (p=0·039). Between-group antiepileptic drugs.35,36 Side-effects decreased progressively
comparisons for 50% responders were not significant during the 3-year follow-up. A compilation of long-term
(23·4% vs 15·7%). However, the high-stimulation group was median change and response rates from three different
more likely to achieve a 75% seizure reduction than the low- studies30,34,37 is shown in figure 3.
stimulation group (10·6% vs 2·0%).
Studies in children with epilepsy
Long-term follow-up studies There have been no controlled trials of VNS in children.
There have been two prospective, long-term, follow-up However, the EO4 study (a prospective open safety study of
studies that assessed the change in seizure frequency and VNS)38 did include 60 children aged 3–18 years. 16 of the
tolerability from the end of the clinical trials for up to 1 year patients were under 12 years of age. In this study, patients with
(EO5 study)30 and up to 3 years (EO1–EO5 studies).34 various types of refractory seizures could be included and
In the EO5 study, 195 patients were followed up for some with primary generalised seizures (27%) were included.
12 months from the time they completed the double-blind After 3 months of stimulation, the median reduction in the
phase and went into the open-label phase of the trial. These number of seizures was 23%; it was 42% at 6 months (n=46),
studies were analysed by three methods: a last-visit carried 34% at 12 months (n=51), and 42% at 18 months (n=46).
forward method, in which the last reported observation was There was no association between improvement and seizure
used at each subsequent time point; a constant-cohort type.
method, in which the patients who reached a particular Another large study with 95 patients was a six-centre
endpoint were analysed at each previous time point; and a retrospective study of efficacy and tolerability of VNS in
declining N method, in which all available patients were children.39 There was an average seizure reduction of 36·1% at
analysed at each time point. Although there are strengths 3 months and 44·7% at 6 months.

THE LANCET Neurology Vol 1 December 2002 http://neurology.thelancet.com 479

For personal use. Only reproduce with permission from The Lancet Publishing Group.
Review Vagus-nerve stimulation

Lennox-Gastaut syndrome were assessed at baseline and again


50 3 and 6 months after implantation. The median age at
E05
45 E03 implantation was 13 years. 60% of patients had IQ scores
40 All VNS studies below 70, 36% had tried the ketogenic diet, and five had
previously had callosotomy. The median seizure reduction
Seizure reduction (%)

35
was 58·2% at 3 months (n=43) and 57·9% at 6 months
30 (n=24). By 6 months, there was a decrease of at least 50% in
25 58% of patients. The falling number of patients in this study
20 was due to study cut-off and not to patients’ dissatisfaction.
The number of atonic seizures was reduced by 55% at
15
3 months and by 88% at 6 months. In the five patients who
10 had had callosotomy, the number of seizures was reduced by
5 73% at 3 months and by 69% at 6 months.44 Other smaller
0 studies have shown similar results 45,46
In conclusion, the efficacy of VNS seems to be established
Patients with >50% seizure reduction (%)

50 for focal-onset seizure types, and there is evidence that it has


45 efficacy for other seizure types as well. Therefore, VNS can be
40 considered to be a broad-spectrum treatment. It seems to be
effective in the most refractory focal-onset seizure patients,
35
even in those who have already had surgical treatment.
30 Improvement is not immediate but tends to increase over
25 18–24 months of treatment. Most of the studies cited above
20 report subjective improvements in various quality-of-life
measures while on VNS. Moreover, recent objective studies
15
have shown improvements in mood,47,48 memory,49 and
10 quality of life.50 A problem is that there have been few
5 prospective and randomised studies, but there is much
0 evidence in open label retrospective studies. One prospective
3 months 1 year 2 years study with a follow-up of 5 years also dealt with various
seizure types and again showed sustained efficacy and
tolerability over time.51
Figure 3. Top: median seizure reduction (%) of patients participating in
E03 (n=114) and E05 (n=105) randomised placebo-controlled trials as Safety and tolerability
well as all VNS studies combined (E01-E05, N=440) at 3 months, 1 year, Acute side-effects
and 2 years of follow-up (intent-to-treat analysis). Bottom: proportion of
Most of the acute complications of VNS implantation have
patients with at least 50% seizure reduction in E03 (N=114) and E05
(N=195) randomised placebo-controlled trials as well as all VNS studies been common infections, vocal-cord pareses, lower facial
combined (E01–E05, N=440) at 3 months, 1 year, and 2 years of follow- weakness, and, in a few cases, bradycardia and asystole.
up (intention-to treat analysis).30,34,37 Postoperative infections occur in 3–6% of patients. Most
are treated with oral antibiotics; rarely are the generator or
VNS and other epileptic disorders electrodes removed.18,30,51 In the EO3 study, infections were not
There have been many case-reports of VNS in different types reported; however, many patients received prophylactic
of epilepsy such as tuberous sclerosis,40 status epilepticus,41 antibiotic treatment. In the EO5 study, infection led to device
hamartoma syndrome,42 infantile spasms, and progressive removal in three of 198 patients (1·5%).18
myoclonic epilepsy of Unverricht-Lundborg type.43 All Paralysis of the left vocal cord occurred in one patient in
reported good efficacy and low frequency of side-effects. the EO3 study but resolved when the device was removed. In
In another six-centre retrospective study, 50 patients with the EO5 study reversible paralysis of the left vocal cord was
reported in two patients.
Proportion of patients with side-effects at stages in
Lower facial weakness was reported in the EO5 study in
long-term treatment
two patients and in one in the EO3 study. This side-effect was
Proportion (%) of patients with side-effects probably due to misplaced surgical incisions when the
Side-effect 3 months18 12 months30 5 years51 electrodes were being connected to the vagus nerve. This side-
effect has become very rare with the improvement of surgical
Cough 21 15 1·5
Voice alteration 62 55 18·7 techniques.
Dyspnoea 16 13 2·3 Ventricular asystole during testing of the device on
Pain 17 15 4·7 implantation has been reported in five patients.52,53 No
Paraesthesia 25 15 1·5 asystolic events have occurred outside the operating room,
Headache 20 16
Pharyngitis 9 10
and patients have been able to use the VNS postoperatively.
Depression 3 5 Similar events have not been reported during device
Infection 4 6 implantation in the clinical trials. The occurrence of this

480 THE LANCET Neurology Vol 1 December 2002 http://neurology.thelancet.com

For personal use. Only reproduce with permission from The Lancet Publishing Group.
Vagus-nerve stimulation
Review

adverse event is estimated to be one in 875 or 0·1%. Possible


reasons could be abnormal electrode placement, indirect Search strategy and selection criteria
Data for this review were identified by searches of Medline,
stimulation of the cervical cardiac nerves, technical
references from the relevant articles, the Cochrane Library,
malfunction of the device, polarity reversal of the leads by the and the files of the author. Search terms included “vagus-
surgeon, or an idiosyncratic reaction. nerve stimulation”, “epilepsy”, “clinical trials”, and “side-
effects”. Only papers published in English were reviewed.
Adverse events related to long-term use Abstracts were used only when no other information was
Most of the data on side-effects are based on studies in adult available.
patients. The most common adverse-side-effects are
stimulus-related coughing, throat pain, and hoarseness, all of replacement of a lead. Hoarseness was reported in 11 patients
which tend to improve with time.34 In the EO3 study the only (18·7%) and throat pain in three (4·7%), Paraesthesia was
side-effects to occur in more than 10% of patients were voice reported in one patient from whom the device and the
alteration (37·2%), hoarseness (37·2%), and throat pain electrodes were removed bacause the side-effect was severe.
(11%). In the EO5 study, side-effects were reported to be Four patients died—three because of status epilepticus, which
more diverse and more common: voice alteration in 66·3% of was caused by infection in two of these patients. The other
patients on high stimulation and 30·0% on low stimulation, patient died of sudden, unexplained death in epilepsy
cough in 45·0% and 42·0%, throat pain in 28·1% on high (SUDEP).
stimulation, dyspnoea in 25·0%, vomiting and paresthesias in
17·9%, and infection in 11·6%.18 The symptoms were mild or Deaths
moderate and rarely required adjustment of the stimulation In 440 patients with refractory epilepsy in the EO1–EO5
variables. No changes in autonomic function were seen in studies, there were nine deaths: four SUDEP one accident,
tests such as blood pressure, heart rate, Holter monitor one thrombocytopenia, one aspiration pneumonia, one renal
measures, lung function, or blood chemistry. failure, and one pneumonia–sepsis. Annegers and
In children, the main side-effects reported are the same as colleagues57 reviewed all deaths in 1819 patients with VNS.
in adults—that is, transient hoarseness, throat pain, and The patients were followed up for 3176·3 person-years from
coughing, all of which are stimulus dependent and tend to implantation and 25 deaths were reported. Rates of SUDEP
improve over time.54,55 Difficulties in swallowing have been were 4·1 per 1000 in patients treated with VNS and 4·5 per
reported in two young children with VNS.54 Examination with 1000 for a control population with refractory epilepsy. After
videoradiography with and without stimulation showed an stratification for duration of VNS use, the prevalence of
increase in aspiration when the device was on. The two SUDEP was 5·5 per 1000 for the first 2 years and then
children, who also had severe motor disabilities, had difficulty dropped to 1·7 per 1000 for the subsequent years. Thus,
in swallowing and needed assisted feeding before VNS. excessive death rates have not been seen in patients with
Schallert and colleagues56 tested swallowing in eight children epilepsy treated with VNS. In fact, there was a tendency for
with VNS in both the on and off phases and did not observe SUDEP rates to be lower than in similar groups of patients
difficulties with aspiration. not treated with VNS.
In conclusion, side-effects are mainly stimulation related
Tolerability and reversible, and they tend to decrease over time. They are
There have been three long-term follow-up reports.30,34,51 195 mild to moderate in most cases and seldom necessitate
patients were included in the 12 month follow-up for the removal of the device. No idiosyncratic side-effects have been
EO5.30 No cardiac events or changes (eg, variability in the reported after 12 years of experience, and VNS does not
intervals between R waves) have been seen. Results of interact with antiepileptic drugs. Most adverse events are
pulmonary function tests and blood-chemistry tests were predictable and related to stimulation variables. CNS side-
unchanged from baseline. 97·8% of the adverse events were effects such as tiredness, psychomotor slowing, irritation, and
reported as mild to moderate and were reversible with a nervousness have not been frequently reported and they do
reduction in stimulation. Noteworthy is the lack of CNS or not stand out as major side-effects of VNS.
cognitive side-effects (table).
In the 3 year follow-up of all patients in EO1–EO5,34 Conclusion
paraesthesias, cough, and hoarseness became less common There is now strong, level I evidence that VNS is an effective
with time. Dyspnoea was the most common adverse event treatment for patients with partial-onset seizures.58,59 There is
reported at 3 years (3·2%). Three serious events of respiratory level II evidence that VNS is effective in most seizure types
difficulties and three of hoarseness were reported, and there from large open-label studies with long-term follow-up
were nine deaths. No changes in Holter monitor or lung- indicating a broad range of activity. Currently, VNS devices
function tests or blood chemistries that could be attributed to are being implanted in patients with refractory seizures who
VNS were noted. cannot have resective surgery or who have had surgery with
In a 5 year follow-up of 64 patients, Ben-Menachem and poor results. Many of these patients have been treated with
co-workers51 reported mainly mild side-effects almost all several antiepileptic drugs before receiving VNS implants.
related to stimulation. One patient complained about device There is, however, preliminary evidence that VNS can be
placement and had it moved twice without satisfaction. One effective in less refractory epilepsy as well,60 and this may be
patient experienced a reversible vocal-cord paresis after the future use of VNS. More research needs to be done to

THE LANCET Neurology Vol 1 December 2002 http://neurology.thelancet.com 481

For personal use. Only reproduce with permission from The Lancet Publishing Group.
Review Vagus-nerve stimulation

provide evidence of efficacy in patients with less refractory with those for antiepileptic drugs. VNS does not have the
disease. VNS, therefore, is now a treatment for cognitive and systemic side-effects seen with the use of drugs
pharmacoresistant epilepsy. Although fewer than 10% of and therefore can be a valuable treatment modality for
patients were reported as seizure-free from the clinical trials, patients who have poor tolerance of effective antiepileptic
this proportion is similar to that seen with antiepileptic drugs drugs.61
in a population with similarly intractable epilepsy.50
Most paediatric neurologists and neurosurgeons also Conflict of interest
agree that VNS should be offered to patients before The author has previously received research grants from Cyebronics,
the company that manufactures vagus-nerve stimulators.
callosotomy because VNS is less invasive and seems to be
effective in patients who would be considered for surgery. Role of the funding source
VNS is well tolerated—side-effects tend to diminish over No funding source was involved in the preparation of this review or the
time, and continuation rates for VNS compare favourably decision to submit it for publications.

References in partial epilepsy. Neurology 1999; 52: 1166–73 hamartomas. Pediatr Neurol 2000; 23: 167–68.
1 Lanska DJ, J.L. Corning and vagal nerve stimulation 24 Henry TR, Bakay RAE, Votaw JR, et al. Blood flow 43 Smith B, Shatz R, Elisevich K, Bespalova IN,
for seizures in the 1880s. Neurology 2002; 58: 452–59. alterations induced by therapeutic vagus nerve Burmeister M. Effects of vagus nerve stimulation on
2 Zabara J. Peripheral control of hypersynchronous stimulation in partial epilepsy 1: acute effects at high progressive myoclonus epilepsy of Unverricht-
discharge in epilepsy. Electroencephalogr Clin and low levels of stimulation. Epilepsia 1998; 39: Lundborg type. Epilepsia 2000; 41: 1046–48.
Neurophysiol 1985; 61: S162 (abstr). 983–90. 44 Frost M, Gates J, Helmers SL, et al. Vagus nerve
3 Zabara J. Time course of seizure control to brief 25 Bohning DE, Lomarev MP, Denslow S, Nahas Z, stimulation in children with refractory seizures
repetitive stimuli. Epilepsia 1985; 26: 518 (abstr). Shastri A, George M. Feasibility of vagus nerve associated with Lennox-Gastaut syndrome. Epilepsia
4 Zabara J. Inhibition of experimental seizures in stimulation-synchronized blood oxygenation level- 2001; 42: 1148–52.
canines by repetitive stimulation. Epilepsia 1992; 33: dependent functional MRI. Invest Radiol 2001; 36: 45 Majorie HJM, Berfelo W, Aldenkamp AP,
1005–12. 470–79. Evers SMAA, Kessels AG, Renier WO. Vagus nerve
5 Bailey P, Bremer F. A sensory cortical representation 26 Ben-Menachem E, Hamberger A, Hedner T, et al. stimulation in children with therapy-resistant epilepsy
of the vagus nerve. J Neurophysiol 1938; 1: 405–12. Effects of vagus nerve stimulation on amino acids and diagnosed as Lennox-Gastaut syndrome: clinical
other metabolites in the CSF of patients with partial results, neuropsychological effects and cost-
6 Zanchetti A, Wang SC, Moruzzi G. The effect of vagal seizures. Epilepsy Res 1995; 20: 221–27. effectiveness. J Clin Neurophysiol 2001; 18: 419–28.
stimulation on the EEG pattern of the cat. EEG Clin
Neurophysiol 1952; 4: 357–461. 27 Naritoku DK, Terry WJ, Helfert RH. Regional 46 Lundgren J, Åmark P, Blennow G, Strömblad LG,
induction of fos immunoreactivity in the brain by Wallstedt L. Vagus nerve stimulation in 16 children
7 Chase MH, Sterman MB, Clemente CD. Cortical and anticonvulsant stimulation of the vagus nerve. Epilepsy with refractory epilepsy. Epilepsia 1998; 39: 809–13.
subcortical patterns of response to afferent vagal Res 1995; 22: 53–62.
stimulation. Exp Neurol 1966; 16: 36–49. 47 Harden CL, Pulver MC, Ravdin LD, Nikolov B,
28 Banzett RB, Guz A, Paydarfar D, et al. Halper JP, Labar DR. A pilot study of mood in
8 Chase MH, Nakamura Y, Clemente CD, Sterman MB. Cardiorespiratory variables and sensation during epilepsy patients treated with vagus nerve stimulation.
Afferent vagal stimulation: neurographic correlates of stimulation of the left vagus in patients with Epilepsy Behav 2000; 1: 93–99.
induced EEG synchronization and desynchronization. epilepsy. Epilepsy Res 1999; 35: 1–11. 48 Elger G, Hoppe C, Falkai P, Rush AJ, Elger CE.
Brain Res 1967; 5: 236–49. 29 Lotvall J, Lunde H, Augustinson LE, Hedner T, Vagus nerve stimulation is associated with mood
9 Stoica I, Tudor I. Effects of vagus afferents on Svedmyr N, Ben-Menachem E. Airway effects of direct improvements in epilepsy patients. Epilepsy Behav
strychnine focus of coronal gyrus. Rev Roum Neurol left-sided cervical vagal stimulation in patients with 2000; 1: 203–10.
1967; 4: 287–95. complex partial seizures. Epilepsy Res 1994; 18: 49 Clark KB, Naritoku DK, Smith DC, Browning RA,
10 Stoica I, Tudor I. Vagal trunk stimulation influences 149–54. Jensen RA. Enhanced recognition memory following
on epileptic spiking focus. Rev Roum Neurol 1968; 5: 30 DeGiorgio CM, Schachter SC, Handforth A, et al. vagus nerve stimulation in human subjects.
203–10. Prospective long-term study of vagus nerve Nat Neurosci 1999; 2: 94–98.
11 Koo B. EEG changes with vagus nerve stimulation. stimulation for the treatment of refractory seizures. 50 Cramer JA. Exploration of changes in health-related
J Clin Neurophysiol 2001; 18: 434–41. Epilepsia 2000; 41: 1195–2000. quality of life after 3 months of vagus nerve
12 Woodbury DM, Woodbury JW. Effects of vagal 31 Krahl SE, Senanayake SS, Handforth A. Destruction of stimulation. Epilepsy Behav 2001; 2: 460-65.
stimulation on experimentally induced seizures in peripheral C-fibers does not alter subsequent vagus 51 Ben-Menachem E, Hellström K, Waldton C,
rats. Epilepsia 1990: 31 (suppl 2): S7–19. nerve stimulation-induced seizure suppression in rats. Augustinsson LE. Evaluation of refractory epilepsy
13 Lockard JS, Congdon WC, DuChare LL. Feasibility Epilepsia 2001; 42: 586–89. treated with vagus nerve stimulation for up to 5 years.
and safety of vagal stimulation: the money model. 32 Krahl SF, Clark KB, Smith DC, et al. Locus coeruleus Neurology 1999; 52: 1265–67.
Epilepsia 1990; 31 (suppl 2): S20–26. lesions suppress the seizure attenuating effects of 52 Tatum WO, Moore DB, Stecker MM, et al.
14 Penry JK, Dean JC. Prevention of intractable partial vagus nerve stimulation. Epilepsia 1998; 39: 709–14. Ventricular asystole during vagus nerve stimulation
seizures by intermittent vagal stimulation in humans: 33 Tarver WB, George RE, Maschino SE. Clinical for epilepsy in humans. Neurology 2000; 52: 1267–69.
preliminary results. Epilepsia 1990; 31 (suppl 2): experience with a helical bipolar stimulating lead. 53 Asconape JJ, Moore DD, Zipes DP, Hartman LM,
S40–43. Pacing Clin Electrophysiol 1992; 15: 1545–56. Duffell WH. Bradycardia and asystole with the use of
15 Uthman BM, Wilder BJ, Hammond EJ, Reid S. 34 Morris GL, Mueller WM, and the Vagus Nerve vagus nerve stimulation for the treatment of epilepsy:
Efficacy and safety of vagus nerve stimulation in Stimulation Study Group EO1–EO5. Long-term a rare complication of intraoperative device testing.
patients with complex partial seizures. Epilepsia 1990; treatment with vagus nerve stimulation in patients Epilepsia 1998; 40: 1452–54.
31(suppl 2): S44–50. with refractory epilepsy. Neurology 1999; 53: 1731–35. 54 Lundgren J, Ekberg O, Olsson R. Aspiration: a
16 Uthman BM, Wilder BJ, Penry JK, et al. Treatment of 35 Krakow K, Walker M, Otoul C, Sander JWAS. Long- potential complication to vagus nerve stimulation.
epilepsy by stimulation of the vagus nerve. Neurology term continuation of levetiracetam in patients with Epilepsia 1998; 39: 998–1000.
1993; 43: 1338–45. refractory epilepsy. Neurology 2001; 56: 1772–74. 55 Hosain S, Nikalov B, Hardin C, Li M, Fraser R,
17 Ben-Menachem E, Manon-Espaillat R, Ristanovic R, 36 Lhatoo SD, Wong ICK, Polizzi G, Sander JWAS. Labar DJ. Vagus nerve stimulation treatment for
et al. Vagus nerve stimulation for treatment of partial Long-term retention rates of lamotrigine, gabapentin, Lennox-Gastaut syndrome. J Child Neurol 2000; 15:
seizures: 1, a controlled study of effect on seizures. and topiramate in chronic epilepsy. Epilepsia 2000; 41: 509–12.
Epilepsia 1994; 35: 616–26. 1592–96. 56 Schallert G, Foster J, Lindquist N, Murphy J. Chronic
18 Handforth A, DeGiorgio CM, Schachter SC, 37 Salinsky GR, Uthman BM, Ristanovic RK, et al. Vagus stimulation of the left vagal nerve in children: effect on
Uthman BM, et al. Vagus nerve stimulation therapy nerve stimulation for the treatment of medically swallowing. Epilepsia 1998; 39: 113–14.
for partial-onset seizures: a randomized active-control intractable seizures: results from a 1-year open- 57 Annegers JF, Coan SP, Hauser WA, Leestma J.
trial. Neurology 1998; 51: 48–55. extension trial. Arch Neurol 1996; 53: 1176–80. Epilepsy, vagal nerve stimulation by the NCP system,
19 Foley JO, DuBois F. Quantitative studies of the vagus 38 Murphy JV and the Pediatric VNS Study Group. Left all-cause mortality, and sudden, unexpected,
nerve in the cat: I, the ratio of sensory to motor fibers. vagal nerve stimulation in children with medically unexplained death. Epilepsia 2000; 41: 549–53.
J Comp Neurol 1937; 67: 49–97. refractory epilepsy. J Pediatr 1999; 134: 563–66. 58 Privitera MD, Welty TE, Ficker DM, Welge J. Vagus
20 Randall WC, Ardell JL. Differential innervation of the 39 Helmers SL, Wheless JW, Frost M, et al. Vagus nerve nerve stimulation for partial seizures (Cochrane
heart. In: Sipes D, Jalife J, eds. Cardiac stimulation therapy in pediatric patients with Review). Chochrane Library 2002: issue 1. CD002896.
electrophysiology and arrhythmias. New York: Grune refractory epilepsy: a retrospective study. J Child 59 Fisher RS, Krauss GL, Ramsay E, Laxer K, Gates J.
and Stratton, 1985: 137–44. Neurol 2001; 16: 843–48. Assessment of vagus nerve stimulation for epilepsy:
21 Rutecki P. Anatomical, physiological, and theoretical 40 Parain D, Penniello MJ, Berquen P, Delangre T, report of the Therapeutics and Technology
basis for the antiepileptic effect of vagus nerve Billard C, Murphy JV. Vagal nerve stimulation in Assessment Subcommittee of the American Academy
stimulation. Epilepsia 1990; 31 (suppl 2): S1–6. tuberous sclerosis complex patients. Pediatr Neurol of Neurology. Neurology 1997; 49: 293–97.
22 Ko D, Heck C, Grafton S, et al. Vagus nerve 2001; 25: 213–16. 60 Wheless J. Interim results of a prospective study of
stimulation activates central nervous system structures 41 Winston KR, Levisohn P, Miller BR, Freeman J. Vagal early adjunctive vagus nerve stimulation (VNS)
in epileptic patients during PET H215O blood flow nerve stimulation for status epilepticus. Pediatr therapy. Neurology 2002; 58 (suppl 3): 1.
imaging. Neurosurgery 1996; 39: 426–30. Neurosurg 2001; 34: 190–92. 61 Cramer J, Ben-Menachem E, French J. Treatment
23 Henry TR, Votaw JR, Bakay RAE, et al. Acute blood 42 Murphy JV, Wheless JW, Schmoll CM. Left vagal options for refractory epilepsy: new medications and
flow changes and efficacy of vagus nerve stimulation nerve stimulation in six patients with hypothalamic vagal nerve stimulation. Epilepsy Res 2001; 47: 17–25.

482 THE LANCET Neurology Vol 1 December 2002 http://neurology.thelancet.com

For personal use. Only reproduce with permission from The Lancet Publishing Group.

S-ar putea să vă placă și