Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011 173
Neuromodulators and its combinations for the
preventive treatment of migraine
Neurotransmissores e suas combinações para o tratamento preventivo
da migrânea
ABSTRACTABSTRACT
ABSTRACTABSTRACT
ABSTRACT
Migraine is a chronic, debilitating neurological disorder. It
affects nearly 15% of the adult population and it is
characterized by a range of symptom profiles and degrees of
disability. It is a disease generally believed to occur in
consequence of a genetically hyper excitable brain state, in
addition to a neurotransmitter dysfunction which results in
susceptibility to the occurrence of intermittent attacks of
headache with particular associated features. Pharmacotherapy
remains the mainstay for the prevention of the attacks and
despite the use of different classes of drugs, some older than
30 years and used by serendipity, some neuromodulators
represent the most modern option and the better studied drugs
for the prophylactic treatment of migraine. Supposedly acting
by targeting one or more molecular sites in the brain, these
drugs alter neurotransmission through effects on ion channels,
on specific receptors and on neurotransmitter metabolism.
Neuromodulators are considered the state of art in migraine
therapeutic and its combination may represent an upcoming
option for patients not responding well or presenting limiting
tolerability issues with full-dose monotherapy. In this review,
we explore the specificities of the different drugs belonging to
this pharmacological class, the evidence available for its use
in migraine as well as the fundamentals and potential for new
approaches combining two neuromodulators, even in lower
doses.
Keywords:Keywords:
Keywords:Keywords:
Keywords: Neuromodulators; Combination; Migraine;
Preventive treatment
VIEW AND REVIEWVIEW AND REVIEW
VIEW AND REVIEWVIEW AND REVIEW
VIEW AND REVIEW
Abouch Valenty Krymchantowski, Carla da Cunha Jevoux
Centro de Avaliação e Tratamento da Dor de Cabeça do Rio de Janeiro
(Headache Center of Rio)
Krymchantowski AV, Jevoux CC
Neuromodulators and its combinations for the preventive treatment of migraine.
Headache Medicine. 2011;2(4):173-81
RESUMORESUMO
RESUMORESUMO
RESUMO
A enxaqueca é uma doença neurológica crônica e incapa-
citante. Afeta em torno de 15% da população adulta e é
caracterizada por vários sintomas e graus diferentes de
incapacidade funcional. A enxaqueca é considerada uma
doença na qual há hiperexcitabilidade cerebral aliada à
disfunção de sistemas de neurotransmissão originando
susceptibilidade à ocorrência de crises intermitentes de cefaleia
com características peculiares. A farmacoterapia preventiva
é o eixo central do tratamento e, a despeito do uso de várias
classes de drogas, algumas com mais de 30 anos e consi-
deradas eficazes por acaso, alguns neuromoduladores repre-
sentam a opção mais moderna e mais estudada para esse
tratamento. Supostamente atuando em um ou mais sítios mole-
culares cerebrais, essas drogas alteram a neurotransmissão
através da ação em canais iônicos, em receptores específicos
ou no metabolismo de neurotransmissores. Os neuromodu-
ladores são considerados o "estado da arte" no tratamento da
enxaqueca e sua combinação pode representar uma opção
nova para pacientes não responsivos ou que apresentam
efeitos colaterais limitando o uso de doses plenas na mono-
terapia com esses fármacos. Nesta revisão, exploramos as
especificidades das diferentes drogas pertencentes a essa
classe, a evidência disponível para sua indicação e funda-
mentos para uma forma nova de utilizá-los através de sua
combinação.
PP
PP
P
alavrasalavras
alavrasalavras
alavras
--
--
-
chave:chave:
chave:chave:
chave: Neuromoduladores; Associação; Migrânia;
Tratamento preventivo
174
Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011
KRYMCHANTOWSKI AV, JEVOUX CC
INTRODUCTION
Migraine is a highly prevalent primary headache,
which affects more women than men and may start during
childhood or adolescence. Those affected may experience
migraine throughout their lives.
(1,2)
Despite its life time
prevalence of 12 to 15% and its disabling nature, migraine
is an underdiagnosed and undertreated disease.
(1)
Migraine is a primary neurological disorder with a clear
genetic basis.
(3,4)
During migraine attacks neural events
result in the dilatation of meningeal blood vessels, which
in turn, results in pain, further nerve activation, and
inflammation.
(5)
It probably results from dysfunction of brainstem
involved in the modulation of craniovascular afferents.
(3-6)
Brainstem activation may also lead to activation of
ascending and descending pathways, with initiation of a
perimeningeal vasodilatation and neurogenic inflammation.
The resulting pain is felt as a combination of altered
perception (due to peripheral or central sensitization) of
stimuli that are usually not painful, as well as the activation
of a feed-forward neurovascular dilator mechanism in the
first division of the trigeminal nerve. Cortical spreading
depression is a presumed substrate of migraine aura;
spreading depression and central dysnociception may also
occur in migraine without aura.
(3-6)
Since the chemical cascade of migraine attacks is
believed to occur, at least in part, consequent to a
genetically hyper excitable brain state, neuromodulators
that decrease neuronal excitability should be effective
approach for the prevention of migrainous
symptoms.
(7-11)
NEUROMODULATORS IN MIGRAINE
Valproate (VLP) is simple, eight-carbon branched-
chain fatty acid with antiepileptic properties, which was
one of the first neuromodulators studied for migraine
prevention. Divalproex (DVP) has also been extensively
studied in controlled-studies. Studies have shown that DVP
decreases migraine headache frequency by 50% or
greater in 45%-50% of the patients after 3 months, versus
12%-15% among those receiving placebo.
(13)
Therefore,
the therapeutic gain of DVP is lower than 35%.
At clinical relevant doses, both VLP and DVP attenuate
plasma protein extravasion in migraine models of
meningeal neurogenic inflammation, and this effect is
reversed by GABA
-A
, but not by GABA
-B
receptor
antagonists.
(14)
Furthermore, the effect of VLP is mimicked
by the GABA
-A
agonist, muscimol, but not by the GABA
-B
agonist baclofen, suggesting a GABA
-A
mediated
mechanism. However, in higher doses it blocks the GABA
degradation by GABA transaminase, thereby increasing
GABA concentrations in both axon and in glial cells. The
role of these pharmacological properties in migraine
prevention is uncertain as it is the DVP action of blocking
voltage-dependent sodium ion-channels, therefore
modulating the release of excitatory amino acids, and of
blocking low-threshold T-type calcium ion channels.
(12)
Although VLP and DVP are more often used in the
preventive treatment of migraine, at least VLP seem also
to be effective for the acute treatment.
(15)
It is established
that the substantia gelatinosa of the spinal cord receives
descending 5-HT fibers from the rostroventral medulla
(RVM) and these fibers connect with spinothalamic
neurons.
(16,17)
Accordingly, VLP action in the acute treatment
of migraine may be partially due to serotoninergic
modulation.
Nowadays, DVP is much more commonly used than
VLP for the preventive treatment of migraine. It is typically
started at a dose of 250 mg bid, and can be brought up
to a dose of 500 mg bid. For the acute treatment, typical
doses range from 300 to 500 mg of intravenous VLP.
Adverse effects limit the use of DVP and include weight
gain, hair loss, potential liver dysfunction, teratogenicity,
among others.
(17)
Topiramate is the most recent medication approved
by the FDA for migraine prevention. It is a sulfamate-
substituted monosaccharide derived from D-fructose that
is structurally distinct from other neuromodulators.(
18)
It has
been proven to be an effective pharmacological agent
at doses ranging from 50 mg to 200 mg/day
(19-23)
(Table
1) for the prevention of migraine and recently for the
treatment of chronic migraine as well.
TPM has modulatory effects on voltage-sensitive
L-type calcium channels.
(12,18)
However, the observation
that TPM is more effective at 10 µM than at 50 µM in
reducing the L-type Calcium currents suggests that TPM
may have a different mode of action from traditional
Calcium channel blockers. The biphasic concentration-
response curve for the effect of TPM on L-type Calcium
currents is similar to that for the modulatory effect of
TPM on GABA
A
receptors.
(24)
Because TPM has no effect
on ionic currents in the absence of GABA, its effect on
GABA
A
receptors appears to be modulatory as well.
(24)
The effect of TPM is similar to that of the benzodiazepines
(BDZs) in that TPM increases the frequency of channel
activation. TPM has been reported to inhibit KA-evoked
Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011 175
whole-cell currents in hippocampal neurons and this is
associated with decrease in neuronal excitability.
(25)
TPM is one of the only neuromodulators associated
with weight loss.
(26,27)
Adverse effects include paresthesias,
cognitive deficits, nephrolithiasis, acute closed angle
glaucoma, and non-anion gap metabolic acidosis-the
latter three considered idiosyncratic in nature. A dose of
50 mg bid has been shown to be optimal, but effects
have been shown at as little as 25 mg bid.
(19,20)
Topiramate's efficacy is similar to the efficacy of DVP,
and it has not been shown to be superior to beta-blockers
or tryciclic anti-depressants, although recent studies have
been suggesting that the combination of topiramate and
other traditional pharmacological agents for migraine
prevention promote better outcome figures for decreasing
the frequency of migraine attacks.
(28-31)
Gabapentin (GBP) is not approved by the FDA for
migraine prevention, but is often used in the treatment of
migraine. Its molecule is formed by the addition of a
cyclohexyl group to GABA, allowing this form of GABA to
cross the blood-brain barrier. It is not metabolized and
does not induce or inhibit hepatic metabolism. Gabapentin
has to be administered three times a day due to its half-
life of 4 to 9 hours and drug-drug interactions are not an
issue with GBP because of its pharmacokinetic profile of
not binding to plasma proteins and its lack of interference
with hepatic function. The mechanism of action of the
gabapentinoids is not fully understood yet. Despite its
structural similarity with GABA, it does not bind to GABA
receptors in the CNS. It does interact with the alfa-2-delta
subunit of voltage-gated ion calcium channels possibly
modulating their currents as well as increases the rate of
GABA synthesis in the brain. Gabapentin has also an
antinociceptive effect. It inhibits monoamine neuro-
transmitter release, including dopamine, serotonin and
noradrenaline in addition to total cellular calcium content.
At the spinal cord level, gabapentin alters N-methyl-D-
aspartate (NMDA) receptor-mediated responses. These
effects explain why GBP has been used in the treatment
of neuropathic pain conditions.
(32-36)
For the prevention of migraine, Gabapentin (1800-
2400 mg/day) was found to be superior to placebo in
reducing the frequency of migraine attacks in a controlled,
double-blind trial, supporting the results of previous open
trials. The responder rate was 36% for gabapentin and
14% for placebo
(37)
(Table 1). The most common adverse
events were dizziness and drowsiness. Clinical experience
does not corroborate the presumed efficacy of gabapentin
and it is not considered one of the neuromodulators
recommended for migraineurs.
Another gabapentinoid, pregabalin, which has a
longer half-life and, therefore, may be used in two-daily
dosages regimen, is also suggested as useful for migraine
prevention despite of the lack of published controlled
studies. Pregabalin is recommended for partial seizures,
pain of post-herpetic neuralgia, pain of the diabetes
mellitus neuropathy, fibromyalgia and generalized anxiety
disorder.
(38,39)
Levetiracetam (LCT) is a pyrrolidine, the racemically pure
S-enantiomer of alfa-ethyl-2-oxo-1-pyrrolidineacetamide.
It inhibits partial and secondarily generalized tonic-clonic
seizures in the kindling model. The mechanisms by which
it exerts this antiseizure effect are still unknown, but despite
its lack of effect on Na
+
channels or either on GABA- or
glutamate mediated synaptic transmission, LCT seems to
act on a binding site at the synaptic vesicle protein SV2A,
at least in rat brain membranes.
(40)
LCT is rapidly and
nearly completely absorbed after oral administration and
it is not bound to plasma proteins; peak serum
concentrations are achieved within 2 hours, and daily doses
are linearly related with plasma concentrations. An
advantage of this neuromodulator is the fact that LCT
NEUROMODULATORS AND ITS COMBINATIONS FOR THE PREVENTIVE TREATMENT OF MIGRAINE
176 Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011
KRYMCHANTOWSKI AV, JEVOUX CC
neither induces nor is a high-affinity substrate for CYP
isoforms or glucuronidation enzymes and thus is devoid
of known interactions with other antiseizure medications,
oral contraceptives or anticoagulants.
(41)
LCT was studied for migraine and chronic migraine
prevention in few trials, mostly uncontrolled. Average dose
was 1,000 mg and results were not impressive, but a better
definition of effective doses in randomized controlled trials
is warranted before this neuromodulator can be excluded
from the migraine medication arsenal.
(42-45)
LCT was also
studied for the prevention of migraine in children. In an
open label prospective trial (n=20), levetiracetam was
used in two daily dosages of 20 mg/kg after an initial
daily dose of 20 mg/kg during one month. In a
retrospective chart review of 19 children, who received
125-700 mg twice daily, migraine frequency was reduced
and headaches attacks were eliminated in 52.6% of the
treated patients.
(46)
Asthenia/somnolence, irritability,
hostility and dizziness were associated with the use of LCT
in this population.
The side effects of LCT reported in initial clinical trials
for epilepsy occurred in at least 3% of the patients and
presented as fatigue or tiredness, somnolence, dizziness
and infection (common cold or upper respiratory tract
infection).
(41,47)
Zonisamide (ZNS), a sulfonamide analog, is a
neuromodulator recently approved as an adjunctive
therapy for partial seizures in adults.
(48)
It has a high oral
bioavailability and a long half-life (63 hours), allowing
therapeutic regimens of once- or twice-daily dosages.
Similarly to topiramate, zonisamide promotes blockade
of voltage-gated sodium channels, inhibition of
potassium-mediated release of glutamate, facilitation of
serotonergic and dopaminergic neurotransmissions and
enhancement of gamma-aminobutyric acid release.
Additionally, it also seems to reduce ion flow through
T-Type calcium channels.
(49,50,51)
ZNS was primarily been tested for the treatment of
refractory migraine. Thirty four patients reported statistically
significant improvement of headache frequency, severity
and duration with a daily dosage of 400 mg/day
(initiation with 100 mg/day and titration till 400 mg/day)
after three months of treatment. Four patients (11.8%)
stopped the treatment due to adverse events, which include
dysphoria and difficulty concentrating.
(47,52)
In a retrospective chart review study of 33 patients
(23 with transformed migraine and 10 with episodic
migraine) who had failed over six preventive drugs prior
to ZNS, an average daily dosage of 340 mg for 6 months
of treatment, provided reduction in the number of
headache days. Adverse events were reported by 14
patients (14.4%), being fatigue the most common.
(53)
Recently, 34 patients with good response to the use
of Topiramate, but interrupting it due to intolerable side
effects, were evaluated after a one-month wash-out period.
Zonisamide was used during 6 consecutive months in a
dose up to 100 mg/day. The mean number of days with
headache per month was reduced from 14,9 ± 5.3 during
the wash-out period to 2,5 ± 0.6 after the treatment
period. Headache severity and disability, as assessed by
visual analog scale and migraine disability assessment
scale, were also significantly reduced. The use of rescue
medications at the end of the study was reduced as well.
Four patients (12%) reported side effects not responsible
for interrupting the treatment.
(51)
Lamotrigine (LTG) is a neuromodulator of the
phenyltriazine class chemically unrelated to existing
neuromodulators. Its chemical structure is 3,5-diamino-
6-(2,3-dichlorophenyl)-as-triazine and has a molecular
formula expressed as C9H7N5Cl2 with a molecular
weight of 256.09. Lamotrigine is very slightly soluble in
water and is well absorbed orally, with up to 98 percent
bioavailability. Absorption is not affected by food.
Approximately 55 percent of the drug is protein bound;
therefore, clinical interaction with other protein-bound
drugs is unlikely. Ninety percent of the drug undergoes
glucuronic acid conjugation in the liver, with the conjugate
and the remaining 10 percent of unmetabolized drug
excreted in the urine.
(54)
The Clearance of LTG is markedly increased by the
co-administration of other antiepileptic drugs that induce
hepatic enzymes. These include carbamazepine,
phenobarbital, phenytoin and primidone. The half-life of
lamotrigine may be reduced by about 50 percent with
concomitant use of one or more of these medications
(Table 2). However, when combined with valproic acid,
its elimination is decreased, and its half-life may be more
than doubled.
(55)
LTG is used as adjunctive therapy or monotherapy
in adults with partial seizures with or without secondary
generalization. The mechanism of action is unknown,
but it stabilizes neural membranes and inhibits the release
of excitatory neural transmitters as glutamate release,
possibly through modulation of voltage-sensitive sodium
channels.
(54)
A role for lamotrigine in the prophylactic treatment
of migraine has been suggested mostly by small open
trials, in which lamotrigine was suggested effective in
Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011 177
reducing the frequency of migraine with aura and aura
symptoms.
(56)
However, a larger double-blind randomized
study demonstrated that lamotrigine was ineffective in
migraine prophylaxis, even after three months of drug
use and more adverse effects were recorded in the
lamotrigine-treated group compared with placebo
(57)
(Table 1). In more recent small, open-label studies, in which
smaller doses were included, lamotrigine was effective in
reducing the frequency of migraine auras and the monthly
rate of migraine with aura attacks.
(58,59,60)
It does
corroborate the importance of larger controlled trials
investigating the true role of lamotrigine in migraine.
Lamotrigine does not impair cognition and the main
contraindication to its use is hypersensitivity to the drug.
The need for monitoring drug levels has not been
established. The most frequently encountered adverse
reactions include dizziness, ataxia, somnolence, headache,
blurred vision, nausea, vomiting and skin rash, which is
seen in approximately 10% of the patients. The risk of
more serious reactions, such as the Stevens-Johnson
syndrome, may be minimized by initializing the drug at a
low dose, escalating it slowly, and avoiding concomitant
use of divalproex or valproate sodium.
(47)
The adamantane derivative memantine (1-amino-
3,5-dimethylaminoadamantane, D-145, Akatinol) (MEM)
is a neuromodulator representing the first in a novel class
of Alzheimer's disease medications acting on the
glutamatergic system. MEM is a moderate-affinity voltage-
dependent noncompetitive antagonist at glutamatergic
N-methyl-D-aspartate (NMDA) receptors.
(61)
By binding
to the NMDA receptor with a higher affinity than
magnesium Magnesium ions, MEM is able to inhibit the
prolonged influx of calcium Calcium ions associated with
neuronal excitotoxicity. In addition, biochemical,
pharmacological, and electrophysiological studies show
that memantine interferes with the metabolism of the
neurotransmitters dopamine, noradrenaline, and
serotonin and modulates synaptic transmission.
(62)
MEM was studied for refractory migraineurs.
Subjects with migraine (episodic migraine with 8-14
days of headache per month or transformed migraine,
who had previously failed at least 2 trials of adequate
preventive therapy) were included. Other preventive
drugs were allowed if the patient had been on a stable
dose for more than 30 days. MEM dose ranged from
10 mg to 20 mg per day and the treatment phase lasted
3 months. The primary endpoint was number of days
with headache at month 3. In the ITT population (n =
28), monthly headache frequency was reduced from
21.8 days at baseline to 16.1 at 3 months (P < .01).
The mean number of days with severe pain was also
reduced from 7.8 to 3.2 at 3 months (P < .01) and
mean disability scores were significantly reduced at 3
months as well, when compared with baseline (36.6 vs
54.9, P < .01). Side effects were present in 37.5% of
the patients; 5.5% dropped out the study because of
poor tolerability. Most adverse events were mild. The
study, although not double-blind, posted preliminary
evidence that MEM could be useful for preventing
refractory migraine.
(62)
EXPERT COMMENTARY
Combining neuromodulators in migraine?Combining neuromodulators in migraine?
Combining neuromodulators in migraine?Combining neuromodulators in migraine?
Combining neuromodulators in migraine?
Managing the migraine patient is sometimes difficult,
especially when they are referred to tertiary centers.
Guidelines recommendations suggest that the goal of
preventive treatment is to reduce headache frequency by
at least 50%, based on the assumption that this reduction
is likely clinically meaningful.
(63-65)
When patients fail to respond as expected to
appropriate therapy, or announces at the first consultation
that he or she has already tried everything and nothing
will work, it is important to identify the reason or reasons
that treatment has failed. Accordingly, although
NEUROMODULATORS AND ITS COMBINATIONS FOR THE PREVENTIVE TREATMENT OF MIGRAINE
178
Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011
KRYMCHANTOWSKI AV, JEVOUX CC
monotherapy is usually recommended, rational
combination therapy is sometimes necessary.
(66,67)
In clinical practice, the use of the neuromodulators
TPM and DVP may be limited by tolerability issues and
optimal doses may not be achieved despite improvement
of headache. Phrases like "This drug helped me with the
headache but I was unable to function" or "I prefer to
keep my headaches and remain thin or with my hair" are
common complaints brought to the health provider
prescribing full doses of these pharmacological
agents.
(68,69)
Clinical experience suggests that patients with good
therapeutic response but poor tolerability may often benefit
from combining medications at smaller doses.
(29,70)
Combining low doses of TPM and DVP may be of interest
also because of their sometimes opposite adverse events
profile (e.g. increase vs. decrease in weight). In addition,
thinking about the fundamentals, specifically regarding
TPM and DVP, one can speculate that a synergistic effect
occurs. Since Valproate increases GABA levels and
potentiates GABA-mediated responses possibly blocking
its degradation by GABA transaminase, and blocks low-
threshold T-type calcium ion channels,
(12,13,17)
whereas
TPM enhances GABA neurotransmission by facilitating
GABA
A
receptor action increasing the opening frequency
of the chloride ion channels in GABA
A
receptors, in
addition to the reduction of the L-type Ca channels
activity, it is reasonable to think that these combined
effects could result in better efficacy on migraine
prevention. Additionally, TPM negatively modulates the
excitatory neurotransmitter glutamate thru binding to the
non-NMDA kainate/AMPA receptors, thereby decreasing
the flow of sodium and calcium ions across the
postsynaptic membrane.
(20,24-26)
In fact, a recent open label trial with a small number
of patients suggested that TPM and DVP, combined in
smaller doses than usually used, was an interesting option
for patients that benefited from therapeutic doses of these
medications but would be otherwise discontinued due to
tolerability issues.
(31)
Another possible approach is the combination of
the modulatory effects of a gabapentinoid, which acts
on alfa-2-delta subunit of voltage-gated ion calcium
channels, modulating their currents and increasing the
rate of GABA synthesis in the brain in addition to alter
N-methyl-D-aspartate (NMDA) receptor-mediated
responses, with TPM, which aims its action also on
calcium channels and glutamatergic system, but in
different receptors.
(12,26,32,33)
Finally, perhaps the potential advantages of obtaining
a modulatory effect of TPM on Kainate/AMPA receptors
with the modulation on NMDA receptors promoted by
memantine also in the excitatory glutamatergic system may
represent an interesting option.
(70)
Although these combinations or any other involving
two neuromodulators have never been tested in
randomized controlled trials, one might speculate on
whether this could be useful for those patients failing the
adequate trials of individual options of this class for
migraine prevention, especially if they needed higher doses
for obtaining efficacy.
Although not every neuromodulator can be combined
with each other due to metabolism interactions and
inductions mediated by inhibition of different types of CYP
enzymes, most of the more recent members of
neuromodulators could be considered as ad on
therapies, for patients not responding or doing so, but
with tolerability issues, when using full doses of a specific
agent (Table 2).
Until it cannot be proved by the rigors of large
controlled studies, the option of combining neuro-
modulators, even in smaller doses, may only be
speculated.
Five-year viewFive-year view
Five-year viewFive-year view
Five-year view
There have been exciting developments in
understanding the molecular biology and involved
mechanisms of migraine in the past years. Since migraine
may involve an unbalance between the excitatory
glutamatergic and inhibitory gabaergic systems as well
as a calcium "channelopathy" directly affecting the
regulation of neurotransmitter release, drugs aiming at
stabilizing the neurochemical synchronization of central
circuits, probably involved in migraine, through actions
on various mechanisms, may, indeed represent powerful
components of the migraine treatment arsenal. However,
as presented, a ceiling effect of 50-60% headache
frequency reduction is the only achieved outcome for most
patients. Additionally, tolerability issues may limit treatment
success due to the impossibility of using full-dose schemes.
Trials on combination therapies for migraine are just
beginning, mostly due to previous lack of funding interest.
Although nothing has been proved yet, especially for the
prevention of migraine, the next few years may represent
a changing paradigm, reasoned by the better outcome
figures obtained with combination of drugs for migraine
acute attacks. The expectations for more efficacious and
Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011 179
better tolerated migraine preventive treatments are
anxiously expected. Until then, exciting results on
combining available drugs may fulfill the upcoming
horizon for relieving the burden of migraine.
Key issuesKey issues
Key issuesKey issues
Key issues
Migraine is a genetically inherited disease, which
involves a brain hiper excitable state
Neurotransmitter dysfunction, probably related to
a calcium channelopathy, is also involved in migraine
The neurotransmitter dysfunction probably results
in a state of central dysnociception and/or dysmodulation
Neuromodulators are effective migraine
preventive pharmacological agents through the decreasing
of neuronal excitability
Some neuromodulators are proven effective.
Others may be used, but further evidence of their efficacy
is still lacking
The combination of two neuromodulators may
useful for some patients who don't tolerate full doses of
individual drugs or need better efficacy outcomes
The future of migraine preventive treatment may
involve two or more drugs aiming at different mechanisms
of action and/or brain circuits
REFERENCES
1. Stewart WF, Schechter A, Lipton RB. Migraine heterogeneity,
disability, pain intensity, and attack frequency and duration.
Neurology. 1994;44(suppl 14):S24-S39.
2. Rasmussen BK. Epidemiology of headache. Cephalagia.
1995;15:45-68)
3. Goadsby PJ. Pathophysiology of migraine. In: Silberstein SD, Lipton
RB, Dalessaio DJ (eds.) Wolff's Headache and Other Head Pain.
7thedition. Oxford University Press, Oxford. 2001:57-72.
4. May A, Goadsby PJ. The trigeminovascular system in humans:
pathophysiology implications for primary headache syndromes
of the neural influences on the cerebral circulation. J Cereb
Blood Flow Metab. 1999;19(2):115-27.
5. Welch KM, Barkley GL, Tepley N, Ramadan NM. Central
neurogenic mechanisms of migraine. Neurology. 1993;43(6
Suppl 3):S21-5.
6. Goadsby PJ, Lipton RB, Ferrari MD. Migraine - current
understanding and treatment. N Engl J Med. 2002;346(4):257-
70. Comment in: N Engl J Med. 2002;347(10):764-6; author
reply 764-6.
7. Ophoff RA, Terwindt GM, Vergouwe MN, van Eijk R, Oefner PJ,
Hoffman SM,, et al. Familial hemiplegic migraine and episodic
ataxia type-2 are caused by mutations in the Ca2+ channel
gene CACNL1A4. Cell. 1996;87(3):543-52.
8. De Fusco M, Marconi R, Silvestri L, Atorino L, Rampoldi L, Morgante
L, et al. Haploinsufficiency of ATP1A2 encoding Na+/K+ pump
alpha2 subunit associated with familial hemiplegic migraine type
2. Nat Genetics. 2003; 33(2):192-6.
9. Welch KM, D'Andrea G, Tepley N, Barkley G, Ramadan NM. The
concept of migraine as a state of central neuronal hyperexcitability.
Neurol Clin. 1990;8(4):817-28.
10. Welch KM. Brain hyperexcitability: the basis for antiepileptic
drugs in migraine prevention. Headache. 2005;45[Suppl 1]:S25-
S32.
11. Mathew NT. Antiepileptic drugs in migraine prevention. Headache.
2001;41[Suppl 1]:S18-S24.
12. Cutrer FM. Antiepileptic drugs: how they work in headache.
Headache. 2001;41[Suppl 1]:S3-S10.
13. Mathew NT, Saper JR, Silberstein SD, Rankin L, Markley HG,
Solomon S, et al. Migraine prophylaxis with divalproex. Arch
Neurol. 1995;52(3):281-6.
14. Tringali G, Aubry JM, Moscianese K, Zamori C, Vairano M,
Preziosi P, et al. Valproic acid inhibits corticotropin-releasing
factor synthesis and release from the rat hypothalamus in vitro:
evidence for the involvement of GABAergic neurotransmission.
J Psychiatry Neurosci. 2004; 29(6):459-66.
15. Mathew NT, Kailasam J, Meadors L, Chernyschev O, Gentry P.
Intravenous valproate sodium (depacon) aborts migraine rapidly:
a preliminary report. Headache. 2000;40(9):720-3.
16. Ramadan NM. Prophylactic migraine therapy: mechanisms and
evidence. Curr Pain Headache Rep. 2004;8(2):91-5.
17. Silberstein SD. Divalproex sodium in headache: literature review
and clinical guidelines. Headache. 1996;36(9):547-55.
18. White HS. Comparative anticonvulsant and mechanistic profile
of the established and newer antiepileptic drugs. Epilepsia. 1999;
40(suppl 5):S2-S10)
19. Silberstein SD, Neto W, Schmitt J, Jacobs D; MIGR-001 Study
Group. Topiramate in migraine prevention: results of a large
controlled trial. Arch Neurol. 2004 Apr;61(4):490-5.
20. Potter DL, Hart DE, Calder CS, et al. A double-blind, randomized,
placebo-controlled, parallel study to determine the efficacy of
topiramate in the prophylactic treatment of migraine. (Abstract)
Neurology 2000;54(suppl 3):A15. *of interest. First study to
demonstrate efficacy of TPM in migraine
21. Cady RK, Schreiber CP, Porter JA, Blumenfeld AM, Farmer KU A
multi-center double-blind pilot comparison of onabotulinumtoxin
A and topiramate for the prophylactic treatment of chronic
migraine. Headache. 2011;51(1):21-32.
22. Valguarnera F, Tanganelli P. The efficacy of withdrawal therapy in
subjects with chronic daily headache and medication overuse
following prophylaxis with topiramate and amitriptyline. Neurol
Sci. 2010;31 (Suppl 1):S175-7.
23. Diener HC, Holle D, Dodick D. Treatment of chronic migraine.
Curr Pain Headache Rep. 2011;15(1):64-9.
24. Sigel E. Functional modulation of ligand-gated GABAA and
NMDA receptor channels by phosphorylation. J Recept Signal
Transduct Res. 1995;15(1-4):325-3.
25. Adelman J, Freitag FG, Lainez M, Shi Y, Ascher S, Mao L, et al.
Analysis of safety and tolerability data obtained from over 1,500
patients receiving topiramate for migraine prevention in controlled
trials. Pain Med. 2008;9(2):175-85.
NEUROMODULATORS AND ITS COMBINATIONS FOR THE PREVENTIVE TREATMENT OF MIGRAINE
180
H
eadache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011
KRYMCHANTOWSKI AV, JEVOUX CC
26. Brandes JL, Saper JR, Diamond M, Couch JR, Lewis DW, Schmitt
J, Neto W, Schwabe S, Jacobs D; MIGR-002 Study Group. JAMA.
2004;291(8):965-73.
27. Krymchantowski A, Tavares C. Weight variations in patients
receiving topiramate migraine prophylaxis in a tertiary care
setting. MedGenMed. 2004;6(3):48.
28. Krymchantowski AV, Jevoux CC. Topiramate vs. divalproex
sodium in the preventive treatment of migraine: a prospective
"real-world" study. Headache. 2011;51:554-558. * of interest.
One first studies to suggest, although not controlled, the similar
efficacy and tolerability of TPM and DVP
29. Pascual J, Rivas MT, Leira R. Testing the combination beta-blocker
plus topiramate in refractory migraine. Acta Neurol Scand.
2007;115(2):81-83. ** study suggesting that combination of a
neuromodulator with a traditional agent is better than the single
use of each drug
30. Keskinbora K, Aydinli I. A double-blind randomized controlled
trial of topiramate and amitriptyline either alone or in combination
for the prevention of migraine. Clin Neurol Neurosurg.
2008;110(10):979-84.
31. Krymchantowski AV, Jevoux CC. Low dose topiramate plus
sodium divalproate for positive responder's intolerant to full
dose monotherapy. Headache. 2011; in press. ** study
suggesting that combining two neuromlodulators, even in lower
doses, may be useful for those not tolerating full doses. May
trigger further controlled studies.
32. Beydoun A, Uthman BM, Sackellares JC. Gabapentin: pharmaco-
kinetics, efficacy and safety. Clin Neuropharmacol. 1995;18(6):
469-81.
33. Kelly KM. Gabapentin: antiepileptic mechanism of action. 1998;
38:139-44.
34. Tremont-Lukats IW, Megeff C, Backonja M-M. Anticonvulsants
for neuropathic pain syndromes: mechanisms of action and place
in therapy. Drugs. 2000;60(5):1029-52.
35. Magnus L. Nonepileptic uses of gabapentin. Epilepsia. 1999;40
(Suppl 6):S66-72; discussion S73-4.
36. Gee NS, Brown JP, Dissanayake VU, Offord J, Thurlow R, Woodruff
GN. The novel anticonvulsant drug, gabapentin (Neurontin),
binds to the alpha2delta subunit of a calcium channel. J Biol
Chem. 1996;271(10):5768-76.
37. Mathew NT, Rapoport A, Saper J, Magnus L, Klapper J, Ramadan
N, et al. Efficacy of gabapentin in migraine prophylaxis. Headache.
2001;41(2):119-28.
38. Tassone DM, Boyce E, Guyer J, Nuzum D. Pregabalin: a novel
gamma-aminobutyric acid analogue in the treatment of
neuropathic pain, partial-onset seizures, and anxiety disorders.
Clin Ther. 2007;29(1):26-48.
39. Arnold L, Mease P, Silverman S. Pregabalin: an alpha-2-delta
ligand for the management of fibromyalgia. Am J Manag Care.
2010;16(5 Suppl):S138-43.
40. Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh
SM, Matagne A, et al. The synaptic vesicle protein SV2A is the
binding site for the antiepileptic drug levetiracetam. Proc Natl
Acad Sci U S A. 2004;101(26):9861-6.
41. McNamara JO. Pharmacotherapy of Epilepsies. In: Brunton L,
Lazo J, Parker K. (eds). Goodman & Gilman's The Pharmacological
Basis of Therapeutics. Eleventh edition. New York: McGraw-Hill,
2006:501-25.
42. Brighina F, Palermo A, Aloisio A, Francolini M, Giglia G, Fierro B.
Levetiracetam in the prophylaxis of migraine with aura: a 6-month
open-label study. Clin Neuropharmacol. 2006; 29(6):338-42.
43. Young W, Shaw J, Bloom M, Gebeline-Myers C. Correlation of
increase in phosphene threshold with reduction of migraine
frequency: observation of levetiracetam-treated subjects.
Headache. 2008;48(10):1490-8. Comment in: Headache.
2009;49(5):783-4.
44. Beran RG, Spira PJ. Levetiracetam in chronic daily headache: a
double-blind, randomised placebo-controlled study. (The
Australian KEPPRA Headache Trial [AUS-KHT]). Cephalalgia.
2011;31(5):530-6.
45. Pizza V, Busillo V, Agresta A, Bisogno A, Capasso A. Elderly
patients with migraine: an open-label study on prophylaxis
therapy with levetiracetam. Cent Nerv Syst Agents Med Chem.
2011;11(1):31-4.
46. Miller GS. Efficacy and safety of levetiracetam in pediatric
migraine. Headache. 2004;44(3):238-43.
47. Kaniecki R. Neuromodulators for migraine prevention. Headache.
2008;48(4):586-600.
48. Frampton JE, Scott LJ. Zonizamide: a review of its use in the
management of partial seizures in epilepsy. CNS Drugs
2005;19(4):347-67. Erratum in: CNS Drugs. 2005;19(7):
633.
49. Biton V. Clinical pharmacology and mechanism of action of
zonisamide. Clin Neuropharmacol. 2007;30(4):230-40.
50. Kito M, Maehara M, Watanabe K. Mechanisms of T-type calcium
channel blockade by zonisamide. Seizure. 1996;5(2):115-9.
51. Villani V, Ciuffoli A, Prosperini L, Sette G. Zonisamide for
migraine prophylaxis in topiramate-intolerant patients: an
observational study. Headache. 2011;51(2):287-91.
52. Drake ME Jr, Greathouse NI, Renner JB, Armentbright AD. Open-
label zonisamide for refractory migraine. Clin Neuropharmacol.
2004;27(6):278-80.
53. Ashkenazi A, Benlifer A, Korenblit J, Silberstein SD. Zonisamide
for migraine prophylaxis in refractory patients. Cephalalgia.
2006;26(10):1199-202.
54. Goa KL, Ross SL, Chrisp P. Lamotrigine: a review of its
pharmacological properties and clinical efficacy in epilepsy.
Drugs. 1993;46(1):152-76.
55. Leach MJ, Marden CM, Miller AA. Pharmacological studies on
lamotrigine, a novel potential antiepileptic drug. II. Neuro-
chemical studies on the mechanism of action. Epilepsia. 1986;
27(5):490-7.
56. Lampl C, Buzath A, Klinger D, Neumann K. Lamotrigine in the
prophylactic treatment of migraine aura- a pilot study.
Cephalalgia. 1999;19(1):58-63.
57. Steiner TJ, Findley LJ, Yuen AW. Lamotrigine versus placebo in
the prophylaxis of migraine with and without aura. Cephalalgia
1997;17(2):109-12. Comment in: Cephalalgia. 1997;17(2):
101-2.
58. D'Andrea G, Granella F, Cadaldini M, Manzoni GC. Effectiveness
of lamotrigine in the prophylaxis of migraine with aura: an open
pilot study. Cephalalgia. 1999;19(1):64-6.
Headache Medicine, v.2, n.4, p.173-181, Oct/Nov/Dec. 2011 181
59. Pascual J, Caminero AB, Mateos V, et al. Lamotrigine in the
prevention of migraine aura [abstract]. Cephalalgia. 2005;
25:646-7.
60. Fumal A, Schoenen J. Effectiveness of lamotrigine in the prophylaxis
of migraine with aura: an open study. [abstract] Cephalalgia.
2006;26:1369-70.
61. Parsons CG, Stöffler A, Danysz W. Memantine: A NMDA receptor
antagonist that improves memory by restoration of homeostasis
in the glutamatergic system: too little activation is bad, too much
is even worse. Neuropharmacology. 2007;53(6):699-723.
62. Bigal M, Rapoport A, Sheftell F, Tepper D, Tepper S. Memantine
in the preventive treatment of refractory migraine. Headache.
2008;48(9):1337-42.
63. Bigal ME, Krymchantowski AV, Lipton RB. Barriers to satisfactory
migraine outcomes. What have we learned, where do we stand?
Headache 2009;49:1028-41. * study discussing the actual
barriers for obtaining success with migraine treatment
64. Headache Classification SubCommittee. The International
Classification of Headache Disorders. 2nd Edition. Cephalalgia.
2004;24(Suppl 1):1-149.
65. Evers S. Treatment of migraine with prophylactic drugs. Expert
Opin Pharmacother. 2008;9(15):2565-73.
66. Krymchantowski AV, Jevoux C da C. Migraine prevention trials
and optimized acute therapy: translating lessons learned into
clinical practice. Curr Pain Headache Rep. 2008;12(3):220-3.
67. Krymchantowski AV, Bigal ME. Polytherapy in the preventive and
acute treatment of migraine: fundamentals for changing the
approach. Expert Rev Neurother. 2006;6(3):283-9.
68. Krymchantowski AV, Bryson J, Lipton RB, Bigal ME. Neuro-
modulators for the treatment of headache disorders and
fibromyalgia. Curr Pain Headache Rep. 2008;12(5):333-7.
69. Krymchantowski AV, Tavares C, Penteado Jd Jde C, Adriano M.
Topiramate in the preventive treatment of migraine: experience
in a tertiary Center. Arq Neuropsiquiatr. 2004;62(1):91-5.
[Article in Portuguese].
70. Pfaffenrath V, Kellhammer U, Pöllmann W. Combination
headache: practical experience with a combination of a beta-
blocker and an antidepressive. Cephalalgia. 1986;6(Suppl
5):25-32.
71. Krymchantowski A, Jevoux C. Memantine in the preventive
treatment for migraine and refractory migraine. Headache.
2009;49(3):481-2.
Correspondence
Abouch VAbouch V
Abouch VAbouch V
Abouch V
alenty Krymchantowski, MDalenty Krymchantowski, MD
alenty Krymchantowski, MDalenty Krymchantowski, MD
alenty Krymchantowski, MD
Headache Center of Rio
Rua Siqueira Campos 43/1002 – Copacabana
22031-070 – Rio de Janeiro, RJ, Brazil
Phone: 55-21-22551055
abouchkrym@uol.com.br
www.dordecabeca.com.br
Received: 10/12/2011
Accepted: 10/23/2011
NEUROMODULATORS AND ITS COMBINATIONS FOR THE PREVENTIVE TREATMENT OF MIGRAINE