You are on page 1of 13

Journal of Neurology (2023) 270:3654–3666

https://doi.org/10.1007/s00415-023-11706-1

NEUROLOGICAL UPDATE

Migraine: from pathophysiology to treatment


Francesca Puledda1   · Elisa Martins Silva2 · Kanokrat Suwanlaong3 · Peter J. Goadsby1,4

Received: 13 March 2023 / Accepted: 3 April 2023 / Published online: 8 April 2023
© The Author(s) 2023

Abstract
Migraine is an extremely disabling, common neurological disorder characterized by a complex neurobiology, involving a
series of central and peripheral nervous system areas and networks. A growing increase in the understanding of migraine
pathophysiology in recent years has facilitated translation of that knowledge into novel treatments, which are currently
becoming available to patients in many parts of the world and are substantially changing the clinical approach to the dis-
ease. In the first part of this review, we will provide an up to date overview of migraine pathophysiology by analyzing the
anatomy and function of the main regions involved in the disease, focusing on how these give rise to the plethora of symp-
toms characterizing the attacks and overall disease. The second part of the paper will discuss the novel therapeutic agents
that have emerged for the treatment of migraine, including molecules targeting calcitonin gene-related peptide (gepants and
monoclonal antibodies), serotonin 5-HT1F receptor agonists (ditans) and non-invasive neuromodulation, as well as providing
a brief overview of new evidence for classic migraine treatments.

Keywords  Migraine · Pathophysiology · Treatment · CGRP · Neuromodulation

Introduction Over the years, our knowledge around migraine has


improved considerably, largely thanks to basic science
Migraine is currently listed as the sixth most disabling dis- and imaging studies allowing us to better understand the
order globally, with the highest ranking among all neuro- complex models that are needed to explain the plethora of
logical disorders [1]. The biology of migraine is complex, migraine symptoms. In fact, pain, the cardinal symptom of
multifactorial and still, for certain aspects, unsolved. The the disorder, is not necessarily the most bothersome for all
underlying feature seems to be a, probably complex, genetic patients at all times [3, 4]. Migraine is characterized by a
predisposition combined with behavioral and environmental succession of key phases that often overlap: the premonitory
conditions that causes an alteration of sensory brain process- (prodromal), aura, pain and postdromal phases [5, 6]. Better
ing, resulting in increased sensory susceptibility. This in turn recognition of these events has allowed us to conceptualize
results in otherwise normal sensory inputs being perceived migraine as a network disorder involving multiple cortical,
as bothersome in migraineurs [2]. subcortical and brainstem regions, generating a wide con-
stellation of signs and symptoms [7]. These areas, which we
* Peter J. Goadsby will analyze in detail in the following sections, have altered
peter.goadsby@kcl.ac.uk function and structure in individuals with migraine and in
animal models of the disease.
1
Headache Group, Wolfson CARD, Institute of Psychiatry, The current review follows a translational and anatomi-
Psychology and Neuroscience, King’s College London,
and National Institute for Health Research (NIHR) SLaM
cal approach, beginning with an outline of the mechanisms
Clinical Research Facility at King’s, Wellcome Foundation and regions that are known to be a part of migraine biology,
Building, King’s College Hospital, London SE5 9PJ, UK before moving on to current acute and preventive treatments,
2
Department of Neurology, Hospital Garcia de Orta, Almada, providing updated references and insights with respect to our
Portugal previous review of 5 years ago [8].
3
Division of Neurology, Department of Medicine, Songkhla
Medical Education Center, Songkhla, Thailand
4
Department of Neurology, University of California, Los
Angeles, Los Angeles, CA, USA

13
Vol:.(1234567890)
Journal of Neurology (2023) 270:3654–3666 3655

Migraine: functional anatomy The role for brainstem regions, such as the PAG and the
and pathophysiology dorsolateral pons in migraine is well established thanks to
observational [23] and neuroimaging studies [24–26], as
The trigeminovascular system and brainstem nuclei well as animal models of migraine showing that the brain-
stem acts as a driver of changes in cortical activity during
The trigeminovascular system consists of peripheral axons migraine [27–30]. Indeed, nuclei such as the LC, rostral
from the trigeminal ganglion that innervate the meninges ventral medulla, superior salivatory and cuneiform nucleus
and intracranial blood vessels peripherally, and converge are key in modulating trigeminovascular pain transmission
centrally in the trigeminocervical complex (TCC), com- and autonomic responses in migraine and represent a site
posed of the spinal trigeminal nucleus caudalis and upper of action for triptans [31, 32], ergot derivatives [33, 34]
cervical spinal cord [9, 10]. Second-order neurons ascend and the novel CGRP receptor antagonists [35, 36]. Further,
from the TCC to thalamocortical neurons and further pro- direct and indirect trigeminal activation of the parabrachial
ject to key brain nuclei in the diencephalon and brain- nucleus may explain why head and facial pain is so intense
stem, such as the locus coeruleus (LC), periaqueductal when compared with noncephalic pain, whereas upward
gray (PAG) and hypothalamus [11, 12]. Activation of the trigemino–parabrachial–limbic connections, particularly
trigeminovascular pain pathways is thought to mediate to the amygdala, can explain affective-motivational aspects
part of the qualities of migraine pain by release of neuro- of migraine and even appetite and taste abnormalities
peptides, such as calcitonin gene-related peptide (CGRP) [12]. Linking the premonitory phase and the onset of pain,
and pituitary adenylate cyclase activating polypeptide neurons in the ventral tegmental parabrachial pigmented
(PACAP), at the level of the dura mater [13–15]. CGRP is ­(VTAPBP) nucleus can modulate trigeminocervical nocic-
widely expressed in both peripheral and central neurons eptive traffic in rat. These effects can be seen with gluta-
and has potent dilatator qualities. It also shows a regu- mate, naratriptan ­(5HT1B/1D receptor agonist), PACAP and
latory action on second- and third-order neurons, which dopamine ­D2/3 mediation [37]. Given the role of the ­VTAPBP
seems to underlie its modulatory role in central pain mech- in hedonic feeding, and the influence of glucose on trigemi-
anisms. CGRP elevation in migraineurs has been linked to nocervical nociceptive transmission [38], the data suggest
a decrease in descending inhibitory mechanisms, which in plausible pathways to explain the much celebrated, yet sel-
turn might lead to migraine susceptibility through sensiti- dom clinically useful, issues of food triggers [39]. One might
zation of multiple central neuronal circuits [13]. re-think some triggers in terms of behaviors arising from
Neurogenic inflammation in the periphery was initially central nervous system activation in the premonitory phase
proposed to be the generator of migraine pain [16], although [40], with very different conclusions concerning cause and
this role has been revisited largely due to the fact that block- effect. Finally, central sensitization of the trigeminovascu-
ers of plasma protein extravasation have failed to treat lar system, especially the trigeminal nucleus caudalis, plays
migraine in clinical trials [17, 18]. While trigeminal acti- an important role in the development of chronic migraine,
vation with associated neurogenic inflammation continues possibly influenced by cytokines release and increased astro-
to be discussed [19], direct evidence for a dural inflamma- cytic activation [41, 42]. Interestingly, as shown by neuro-
tory component in migraine is lacking. As described above, physiological [43] and neuroimaging studies [44], brainstem
migraine is associated with a spectrum of sensory dysfunc- activations seems most prominent in the 24 h preceding the
tions with cycling behavior, during which the headache headache onset, and declines during the attack.
phase represents the plateau of trigeminal nociceptive acti-
vation [20]. One hypothesis is that peripheral trigeminovas- The hypothalamus
cular neurons are sensitized, and thereafter sensitize second-
order neurons in the trigeminal nucleus caudalis and upper The theories on migraine as a cyclic sensory threshold disor-
cervical spinal cord, and project rostrally to thalamic nuclei der have highlighted the importance of the hypothalamus as
and key medullary, brainstem and diencephalic regions [21]. a central facilitator of pain, and also of the constellation of
Studies have reliably provided evidence for early brainstem premonitory symptoms such as yawning, thirst and polyuria
involvement in the nociceptive migraine phase; however, it [45], which can precede and continue into the pain phase.
is becoming clearer that the initiation of a migraine attack Functional neuroimaging performed during spontaneous and
is linked to intrinsic brain dysfunction in more central areas nitroglycerin-triggered attacks has consolidated the role of
such as the hypothalamus, and possibly to external factors the hypothalamus in migraine initiation [46]. Altered hypo-
as well [22]. Whether the central dysfunction, very clearly thalamic-brainstem connectivity with the spinal trigeminal
demonstrated in the premonitory phase, facilitates central nuclei and the dorsal rostral pons has been shown in the
sensitization remains an intriguing area of study. premonitory phase of migraine [44] for up to 48 h preced-
ing pain onset [47]. Positron emission tomography has also

13

3656 Journal of Neurology (2023) 270:3654–3666

previously revealed hypothalamic activation during both chronic migraine patients have greater activation of ascend-
spontaneous migraine headache [48] and the premonitory ing trigeminal somatosensory pathways and lower activation
phase [49]. Recent functional imaging studies have even of top-down pain modulatory circuits. This could indicate
shown that changes in hypothalamic connectivity with the interictal dysfunction of the descending pain modulatory
spinal trigeminal nucleus and cortical regions are associated system and amplification of nociceptive processing in
with the development of chronic migraine [50, 51]. migraineurs, mediated by the thalamus and possibly con-
The mechanism(s) by which the hypothalamus can tributing to central sensitization [72].
become ‘overactive’ in migraine, leading to sensitization The processing of trigeminovascular nociceptive informa-
of trigeminal nociceptors is still unclear. Anatomically, the tion in the thalamus can represent a target for management,
hypothalamus has direct and indirect connections to the thal- and indeed several migraine treatments including triptans
amus [52], the trigeminovascular system [53] and to sym- [31, 73], preventives [74–77] and non-invasive neuromodu-
pathetic and parasympathetic brainstem neurons [54], influ- lation have been shown to modulate thalamocortical activity
encing nociceptive and autonomic regulation in migraine. [78, 79].
Stress, which is said to be a common trigger of migraine,
can activate the kappa opioid receptor on tuberoinfundibular The cortex
dopaminergic neurons and lead to an increase in circulat-
ing prolactin causing sensitization of trigeminal afferents, The role of the cerebral cortex in migraine was initially
particularly in females [55]. Further, the hypothalamus has linked to the aura phenomenon and its peculiar symptoms
chemosensitive neurons that can detect metabolic changes in [80, 81]. Aura is thought to be generated by cortical spread-
the brain and periphery. Exogenous stimuli causing a change ing depression (CSD) [82], which has been indirectly evi-
in homeostasis and this intrinsic biorhythm could thus pos- denced in humans through functional neuroimaging [83].
sibly ‘tip’ the brain towards a migraine attack via activation Although CSD can activate the trigeminovascular system in
of the hypothalamus [56]. animals [84, 85] possibly through CGRP-mediated mecha-
nisms [86], it is unlikely to contribute to the headache and
The thalamus possibly constitutes an epiphenomenon of migraine [87, 88].
Regardless of CSD, the cortex has been increasingly
The thalamus has a critical role in sensory processing, implicated in migraine genesis, and in fact many changes in
receiving inputs from the extracranial skin and dura mater the structure and function of key cortical areas associated
from second-order trigeminovascular neurons, and project- with pain processing have been reported in patients, both
ing to cortical regions involved in autonomic, affective, and in the ictal and interictal period [89]. During the headache
cognitive functions—all of which explains in part the com- phase, cortical networks including the salience, sensorimo-
plexity of migraine features [57]. Thalamocortical synchro- tor, default mode, executive and attentional networks, show
nization is affected by a network of neurotransmitters and functional changes; this reflects the cognitive, painful and
neuropeptides in the brainstem (glutamate, serotonin, and emotional symptoms of migraine [90]. Studies in patents
noradrenaline), reticular thalamic regions (γ-aminobutyric with aura have consistently shown differences in brain struc-
acid—GABA) and hypothalamic nuclei (dopamine, hista- ture [91, 92], functional connectivity [92], cortical excit-
mine, orexin, and melanin-containing hormone [58]). There ability [93–95] and pain modulation in the visual pathways
is abundant clinical and preclinical evidence showing that [96]. Occipital cortex involvement in particular can explain
the thalamus is crucial for the development of central sen- the plethora of visual symptoms associated with migraine,
sitization, photophobia and allodynia in migraine [59–64]. from light sensitivity to visual aura and visual snow [97].
Structural neuroimaging studies have shown differences Menstrual migraine has recently been linked to structural
in volume of thalamic nuclei with microstructural abnor- and functional connectivity changes in the right anterior cin-
malities [65–67]. However, such changes were not seen gulum [98] an area involved in the cognitive processing of
in a recent large study involving female patients with aura pain and previously associated with migraine biology [99].
[68]. Functional MRI studies have also shown important However, evidence from neuroimaging studies has been
changes in the thalamus, both within and outside of attacks. inconclusive at times [100], with meta-regression analyses
In migraine without aura, connectivity between the thala- failing to pinpoint alterations that are specific to migraine
mus and pain modulating areas seems to be affected during [101], showing that further research on the topic is needed.
the ictal phase [69]. Abnormal low-frequency oscillations Of note, the association of white matter hyperintensity
in dynamic thalamocortical networks  are implicated in (WMH) in migraine has long been debated [102], particu-
the interictal phase [70], with changes in pulvinar activity larly in migraine with aura [103, 104]. Recently, an asso-
allowing differentiation between migraineurs and controls ciation was identified between the presence of juxtacortical
[71]. Another recent study showed that both episodic and WMHs within the frontal lobe with patient age and duration

13
Journal of Neurology (2023) 270:3654–3666 3657

of disease [105]. WMHs have also been associated with nau- 5-HT1B and 5-HT1D [119], which inhibit CGRP release
sea, vomiting, dizziness and pain intensity during attacks, [120]. The class includes seven options in different formu-
[106]. lations [121], which can be switched to find the optimal
Dysfunctional cortical mechanisms and in particular combination for efficacy and tolerability in the individual
thalamocortical dysrhythmia have also been implicated in patient and which can be combined with NSAIDs to prolong
the mechanism underlying the lack of habituation typical of therapeutic effect and limit rebounds [122, 123]. There are
migraine [107]; in this, repeated sensory stimuli cause an gender-related differences in triptan tolerability, as women
incremental, instead of a reduction, increase in the ampli- seem to present higher adverse event frequency and head-
tudes of sensory responses [108, 109]. Lack of habituation, ache recurrence rates with these drugs [124].
measured for different sensory modalities, usually occurs Non-responsiveness to triptans may be categorized into
during the pain-free period and reverts during the ictal phase refractory: failure of three triptans, one of which should be
or when attacks become more frequent [110]. subcutaneous sumatriptan; and resistant: failure of at least
Finally, large genome-wide association studies (GWAS) two triptans [125]. Non-responsiveness can have a signifi-
have identified susceptibility gene variants in migraine cant impact on health-related quality of life and work pro-
patients, mostly involved in glutamatergic neurotransmis- ductivity [126] and has been linked by recent neuroimaging
sion, which could lead to abnormal cortical excitability and data to changes in hippocampal volume [127]. Importantly,
altered plasticity [111], as evidenced by numerous magnetic even if clinical practice has not demonstrated strong drug-
resonance spectroscopy studies performed over the years related cardiovascular risk [128], triptans are still con-
[112]. Readers interested in the genetics are referred to this traindicated in at-risk patients due to their vasoconstrictive
recent article [113]. Given the complexity that has emerged qualities [129]. The new classes of ditans and gepants do not
from GWAS work, in which each change has such a modest present this disadvantage.
effect on the overall phenotype, one might reflect on whether
clinically useful genetic changes will emerge in the near Ditans
term that will have an impact on management and treatment.
Lasmiditan is the only ditan currently available; it is a
potent and selective 5-HT1F receptor agonist [130], acting
Novel therapies in migraine in migraine by blocking activation of neurons in the trigemi-
nal nucleus caudalis [131] without affecting the vasculature
The last few years have represented an exciting and promis- [130]. Lasmiditan has now been studied in two-phase two
ing time in the field of migraine, thanks to the introduction studies [132, 133] and three large phase three randomized
of several new medications in clinical practice, and with controlled trials [134–136] and shown to have better efficacy
other therapeutic targets, such as glutamate, amylin, adre- compared to placebo on rates of 2-h pain freedom and free-
nomedullin, orexins and pituitary adenylate cyclase activat- dom from most bothersome symptoms, particularly with the
ing polypeptide, currently all in the therapeutic pipeline 100 and 200 mg doses. Pooled data from the phase 3 studies
[114, 115]. The new novel treatments have rapidly changed showed no cardiovascular safety concerns, and in fact, these
the paradigm of migraine management, particularly chal- included patients with coronary artery disease, complicated
lenging the dichotomous division between acute and pre- cardiac arrhythmias and/or hypertension [137]. Across these
ventive medication, which we will however, follow in this studies, neurological side effects—particularly dizziness,
review for simplicity. Further, patient-reported outcomes nausea and somnolence—were common, but mostly mild
such as interictal burden and time lost due to an attack are to moderate and self-limiting [138].
becoming more relevant in the consideration of efficacy and
tolerability of these drugs [116, 117], allowing for signifi- Gepants
cant advances in the management of this condition [118].
Gepants are small-molecule CGRP receptor antagonists
Acute treatments developed for use in the acute treatment of migraine. Six
gepants were initially developed for acute use in migraine,
Therapy for migraine attacks includes non-steroidal anti- with two being discontinued due to liver toxicity [139, 140],
inflammatory drugs (NSAIDs), combination analgesics, one due to lack of oral availability [141] and one for com-
ergotamine preparations and migraine-specific medications. mercial reasons [142]. Ubrogepant and rimegepant repre-
The latter class, which until a few years ago meant triptans, sent a new generation of oral gepants that have received
has recently grown to include ditans, serotonin ­5 HT1F FDA approval for acute migraine therapy [143, 144] fol-
receptor agonists, and gepants, CGRP receptor antagonists. lowing phase 3 studies: Achieve I [145] and II [146] for
Triptans are full agonists of presynaptic serotonin receptors ubrogepant, and Study 301 [147], 302 [148] and 303 [149]

13

3658 Journal of Neurology (2023) 270:3654–3666

for rimegepant. Ubrogepant has been approved at 50 and option for migraine prevention, with treatment to be con-
100 mg doses and Rimegepant is available in an orodis- tinued as long as needed [181], although in most jurisdic-
persible (lyophilized) form at a dose of 75 mg. Preliminary tions, this is not possible to operationalize easily.
evidence also shows effectiveness of the zavegepant nasal Two gepants, atogepant and rimegepant, have recently
spray, a non-oral gepant [150]. Importantly, gepants do not been introduced into the market after proving effective
seem to cause medication overuse headache, making them and well tolerated for the preventive treatment of migraine
a useful option when managing this complication [151] and [182–184]. Both have a similar short half-life of around
can be taken in multiple doses during the attack with good 11 h [185, 186], which facilitates their preventive indica-
rates of success [152]. The low side effect profile of gepants tion. They bring the advantage of fewer adverse events
is appealing; however, caution in the early days of real-world and increased safety, particularly in women who have
use is merited [153]. unplanned pregnancies given the difference in half-life
Metabotropic and ionotropic glutamate receptors may compared to monoclonal antibodies [151]. Atogepant was
become important targets in the future acute therapy of directly designed as a preventive agent, and has been FDA-
migraine, although adverse event issues need to be over- approved for episodic migraine [187]. The most common
come. Experimental and clinical studies have shown an side effects in the phase 2b and 3 studies were constipation
effect of NMDA, AMPA, iGluR5 and mGluR5 recep- and nausea, each at 10% at the 60 mg daily dose. The cur-
tor antagonists in migraine, although their efficacy was rently available data suggests safety on cardiac repolari-
lower than that of sumatriptan and visual side effects were zation even with supratherapeutic doses and, in contrast
observed [154–156]. Blockers of the metabotropic glutamate with the first gepants, no elevation of serum alanine ami-
receptor 5 in particular, or glurants, have a strong clinical notransferase [188, 189]. A further study for the preven-
potential for becoming a candidate drug class for migraine, tive treatment in chronic migraine has been reported in
if the relevant issues of hepatoxicity and transient dizziness abstract form [190]. It is being studied in combination with
can be resolved [157]. The NMDA receptor is also relevant onabotulinumtoxinA (NCT05216263) and for long-term
for migraine with aura, as evidenced by a small RCT show- safety and tolerability in another trial (NCT04686136).
ing the efficacy of ketamine on reducing the severity of auras Rimegepant, initially trialed for acute use, can prevent
[158]. episodic migraine in adults when taken every other day
[184] with the additional benefit of it being used concomi-
tantly during a migraine attack. It is well tolerated, with
Preventive treatments nausea occurring in 2% of cases and this is the most com-
mon side effect.
Preventive therapy is recommended in patients who are New evidence on efficacy and tolerability has also
affected by migraine on at least 2 days per month, when emerged for well-known migraine preventives. A meta-
there is medication overuse and/or when quality of life is analysis has documented the efficacy in chronic migraine
impaired [159]. The application of this guidance will be of OnabotulinumtoxinA, which allows for a reduction of
governed by clinical judgment in the individual case. Clas- over 50% in migraine days after 24 weeks of treatment
sic prevention includes different drug categories, such as [191]. Several open-label studies have also shown a benefit
β blockers, anticonvulsants, tricyclic antidepressants and of combining onabotulinumtoxinA with mAbs to CGRP
calcium channel modulators, which, however, often lead for CM [192–194]. A recent head-to-head trial for chronic
to tolerability issues and poor compliance [160]. In recent migraine showed non-inferiority between propranolol and
years, monoclonal antibodies (mABs) against the CGRP topiramate, with no significant difference in adverse events
peptide (galcanezumab, fremanezumab, eptinezumab) or incidence between the two [195]. Another prospective ran-
its canonical receptor (erenunmab) have been widely intro- domized trial in chronic migraine compared flunarizine
duced in clinical practice and treatment guidelines [161]. 10 mg with topiramate 50 mg daily, showing both drugs
These treatments have persistently confirmed their efficacy had a similar safety profile, with flunarizine being overall
in phase 3 trials [162–174], with convenient dosing, faster more effective [196]. Recent retrospective studies have
onset of efficacy and mild to moderate adverse events [175, confirmed the usefulness of candesartan as a first-line
176]. Further, real-world studies have shown improvement migraine preventive, even in patients who failed numer-
with mABs and worsening of migraine frequency follow- ous previous drugs [197, 198]. Finally, meta-analyses have
ing discontinuation, with most patients resuming treatment been conducted on melatonin [199, 200] and memantine
as soon as possible following breaks due to regulatory [201], both showing favorable side effect profiles and good
restrictions [177–180]. The European Headache Federa- efficacy in migraine.
tion currently recommends CGRP mABs as a first-line

13
Journal of Neurology (2023) 270:3654–3666 3659

Neuromodulation study showed preliminary evidence supporting its use in


chronic migraine [222]. Finally, repetitive peripheral mag-
Non-invasive neuromodulation is an evolving field and is netic stimulation (rPMS) targeting the muscles in the neck
of particular clinical interest for migraine management as and shoulder muscles has shown efficacy in the prevention of
it offers the option of being used both as an acute and pre- episodic migraine, particularly in patients with a high level
ventive treatment. It also presents near to no systemic side of muscular involvement [223, 224].
effects and can thus be offered to patients that present toler- Although these techniques are promising for the man-
ability issues or who need to avoid medication interaction agement of a disabling condition with often little treatment
[202]. options, further evidence is needed to evaluate the scope of
The devices used in migraine target the nervous system their effect in migraine, including novel mechanisms and
through a transcutaneous approach, either centrally (single- targets [225, 226].
pulse transcranial magnetic stimulation, or sTMS) or in the
periphery (non-invasive vagus nerve stimulation or nVNS,
supraorbital nerve stimulation or SNS and transcranial direct
current stimulation or tDCS). Conclusions
A handheld sTMS device is now approved in the USA and
Europe for the acute and preventive treatment of migraine, The last 2 decades have been an incredibly exciting period
following positive results as an acute migraine treatment in for clinicians and researchers interested in migraine, as they
a RCT involving 164 migraineurs with aura [203] and sub- have seen a rapid increase in studies that have led to a greater
sequent post-marketing survey [204] and open-label study knowledge and understanding of the neurobiology of the
[205] demonstrating an effect on headache day reduction disorder. From suffering with a condition that was often
and 50% responder rate. The efficacy of sTMS in migraine overlooked and under-managed, migraineurs are now being
prevention has also been shown in difficult-to-treat patients offered novel treatments that are more and more tailored
[206]. to their needs, and that are fundamentally re-shaping our
External trigeminal (supraorbital) nerve stimulation has approach to the disease.
also shown promise with supporting evidence for the treat- More research and progress is needed and is expected in
ment of migraine, with one RCT showing higher efficacy the coming years, and hopefully this will continue to raise
and tolerability than sham in 109 patients after 1 h of acute awareness around a complex phenomenon affecting millions
treatment [207]. For prevention, its effect seems greater in of people all over the world.
episodic migraine [208] than in refractory [209] or chronic
migraine patients [210].
Open Access  This article is licensed under a Creative Commons Attri-
Regarding non-invasive vagus nerve stimulation bution 4.0 International License, which permits use, sharing, adapta-
(nVNS), it has shown evidence of efficacy in a RCT for the tion, distribution and reproduction in any medium or format, as long
acute treatment of migraine [211], but not for prevention as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
[212–214]. From a mechanistic perspective, this approach at
were made. The images or other third party material in this article are
the bench can suppress cortical spreading depression [215] included in the article's Creative Commons licence, unless indicated
and inhibit trigeminocervical neurons responding to duro- otherwise in a credit line to the material. If material is not included in
vascular nociceptive activation [216]. the article's Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
Another approach has focused on the application of
need to obtain permission directly from the copyright holder. To view a
repeated cathodal or anodal transcranial direct current stimu- copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/.
lation over the cortex, although data on its therapeutic effect
in migraineurs has been conflicting [217, 218]. This may be
due to methodological differences regarding the techniques,
the targeted brain regions and stimulation types [219], war- References
ranting further investigation.
Novel options for neuromodulation include the remote 1. Global Burden of Disease Study 2013 Collaborators (2013)
noncephalic electrical neurostimulation of the upper arm Global, regional, and national incidence, prevalence, and years
lived with disability for 301 acute and chronic diseases and inju-
skin. The device works non-invasively through conditioned ries in 188 countries, 1990–2013: a systematic analysis for the
pain modulation and [220] has been evaluated in a RCT Global Burden of Disease Study 2013. Lancet (London, England)
involving 253 patients. Participants reported clinically 386(9995):743–800
meaningful relief from migraine pain and pain freedom after 2. Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J,
Schankin C, Akerman S (2017) Pathophysiology of migraine—
2 h of treatment compared to sham, with a low incidence a disorder of sensory processing. Physiol Rev 97(2):553–622
of device-related adverse events [221]. A recent open-label

13

3660 Journal of Neurology (2023) 270:3654–3666

3. Lipton RB, Dodick DW, Ailani J, McGill L, Hirman J, Cady 26. Stankewitz A, Aderjan D, Eippert F, May A (2011) Trigemi-
R (2021) Patient-identified most bothersome symptom in pre- nal nociceptive transmission in migraineurs predicts migraine
ventive migraine treatment with eptinezumab: a novel patient- attacks. J Neurosci 31(6):1937–1943
centered outcome. Headache 61(5):766–776 27. Goadsby PJ, Duckworth JW (1989) Low frequency stimulation
4. Lampl C, Thomas H, Stovner LJ, Tassorelli C, Katsarava Z, of the locus coeruleus reduces regional cerebral blood flow in the
Laínez JM et al (2016) Interictal burden attributable to episodic spinalized cat. Brain Res 476(1):71–77
headache: findings from the Eurolight project. J Headache Pain 28. Vinogradova LV (2015) Comparative potency of sensory-induced
17:9 brainstem activation to trigger spreading depression and seizures
5. Dodick DW (2018) A Phase-by-phase review of migraine patho- in the cortex of awake rats: implications for the pathophysiology
physiology. Headache 58(Suppl 1):4–16 of migraine aura. Cephalalgia 35(11):979–986
6. Karsan N, Goadsby PJ (2018) Biological insights from 29. Knight YE, Bartsch T, Kaube H, Goadsby PJ (2002) P/Q-type
the premonitory symptoms of migraine. Nat Rev Neurol calcium-channel blockade in the periaqueductal gray facilitates
14(12):699–710 trigeminal nociception: a functional genetic link for migraine? J
7. Charles A (2013) Migraine: a brain state. Curr Opin Neurol Neurosci 22(5):Rc213
26(3):235–239 30. Knight YE, Goadsby PJ (2001) The periaqueductal grey matter
8. Puledda F, Messina R, Goadsby PJ (2017) An update on modulates trigeminovascular input: a role in migraine? Neurosci-
migraine: current understanding and future directions. J Neu- ence 106(4):793–800
rol 264(9):2031–2039 31. Kroger IL, May A (2015) Triptan-induced disruption of
9. Goadsby PJ, Hoskin KL (1997) The distribution of trigemi- trigemino-cortical connectivity. Neurology 84(21):2124–2131
novascular afferents in the nonhuman primate brain Macaca 32. Goadsby PJ, Hoskin KL (1996) Inhibition of trigeminal neurons
nemestrina: a c-fos immunocytochemical study. J Anat 190(Pt by intravenous administration of the serotonin (5HT)1B/D recep-
3):367–375 tor agonist zolmitriptan (311C90): are brain stem sites therapeu-
10. Hoskin KL, Zagami A, Goadsby PJ (1999) Stimulation of the tic target in migraine? Pain 67(2–3):355–359
middle meningeal artery leads to Fos expression in the trigemi- 33. Goadsby PJ, Gundlach AL (1991) Localization of 3H-dihydro-
nocervical nucleus: a comparative study of monkey and cat. J ergotamine-binding sites in the cat central nervous system: rel-
Anat 194:579–588 evance to migraine. Ann Neurol 29(1):91–94
11. Akerman S, Holland PR, Goadsby PJ (2011) Diencephalic 34. Hoskin KL, Kaube H, Goadsby PJ (1996) Central activation of
and brainstem mechanisms in migraine. Nat Rev Neurosci the trigeminovascular pathway in the cat is inhibited by dihy-
12(10):570–584 droergotamine. A c-Fos and electrophysiological study. Brain J
12. Goadsby PJ, Holland PR (2019) An update: pathophysiology Neurol 119(Pt 1):249–256
of migraine. Neurol Clin 37(4):651–671 35. Pozo-Rosich P, Storer RJ, Charbit AR, Goadsby PJ (2015)
13. Ho TW, Edvinsson L, Goadsby PJ (2010) CGRP and its recep- Periaqueductal gray calcitonin gene-related peptide modulates
tors provide new insights into migraine pathophysiology. Nat trigeminovascular neurons. Cephalalgia 35(14):1298–1307
Rev Neurol 6(10):573–582 36. Storer RJ, Akerman S, Goadsby PJ (2004) Calcitonin gene-
14. Iyengar S, Johnson KW, Ossipov MH, Aurora SK (2019) related peptide (CGRP) modulates nociceptive trigeminovascular
CGRP and the trigeminal system in migraine. Headache transmission in the cat. Br J Pharmacol 142(7):1171–1181
59(5):659–681 37. Martins-Oliveira M, Akerman S, Holland PR, Tavares I, Goadsby
15. Zagami AS, Edvinsson L, Goadsby PJ (2014) Pituitary adenylate PJ (2022) Pharmacological modulation of ventral tegmental area
cyclase activating polypeptide and migraine. Ann Clin Transl neurons elicits changes in trigeminovascular sensory process-
Neurol 1(12):1036–1040 ing and is accompanied by glycemic changes: Implications for
16. Markowitz S, Saito K, Moskowitz MA (1987) Neurogenically migraine. Cephalalgia Int J Headache 42(13):1359–1374
mediated leakage of plasma proteins occurs from blood vessels 38. Martins-Oliveira M, Akerman S, Holland PR, Hoffmann JR,
in dura mater but not brain. J Neurosci 7:4129–4136 Tavares I, Goadsby PJ (2017) Neuroendocrine signaling modu-
17. May A, Goadsby PJ (2001) Substance P receptor antagonists in lates specific neural networks relevant to migraine. Neurobiol
the therapy of migraine. Expert Opin Investig Drugs 10:1–6 Dis 101:16–26
18. Peroutka SJ (2005) Neurogenic inflammation and migraine: 39. Martins-Oliveira M, Tavares I, Goadsby PJ (2021) Was it
implications for therapeutics. Mol Interv 5:306–313 something I ate? Understanding the bidirectional interaction of
19. Ramachandran R (2018) Neurogenic inflammation and its role migraine and appetite neural circuits. Brain Res 1770:147629
in migraine. Semin Immunopathol 40(3):301–314 40. Karsan N, Bose P, Newman J, Goadsby PJ (2021) Are some
20. Peng KP, May A, Basedau H (2022) Cycling multisensory patient-perceived migraine triggers simply early manifestations
changes in migraine: more than a headache. Curr Opin Neurol of the attack? J Neurol 268(5):1885–1893
35(3):367–372 41. Edvinsson L, Haanes KA, Warfvinge K (2019) Does inflamma-
21. Harriott AM, Orlova Y (2022) Anatomy and physiology of head- tion have a role in migraine? Nat Rev Neurol 15(8):483–490
ache. Semin Neurol 42(4):459–473 42. Zhang L, Lu C, Kang L, Li Y, Tang W, Zhao D et al (2022) Tem-
22. Messina R, Gollion C, Christensen RH, Amin FM (2022) Func- poral characteristics of astrocytic activation in the TNC in a mice
tional MRI in migraine. Curr Opin Neurol 35(3):328–335 model of pain induced by recurrent dural infusion of inflamma-
23. Raskin NH, Hosobuchi Y, Lamb S (1987) Headache may arise tory soup. J Headache Pain 23(1):8
from perturbation of brain. Headache 27(8):416–420 43. Hsiao F-J, Chen W-T, Pan L-LH, Liu H-Y, Wang Y-F, Chen S-P
24. Weiller C, May A, Limmroth V, Juptner M, Kaube H, Schayck et al (2022) Dynamic brainstem and somatosensory cortical
RV et al (1995) Brain stem activation in spontaneous human excitability during migraine cycles. J Headache Pain 23(1):21
migraine attacks. Nat Med 1(7):658–660 44. Schulte LH, May A (2016) The migraine generator revisited:
25. Bahra A, Matharu MS, Buchel C, Frackowiak RS, Goadsby PJ continuous scanning of the migraine cycle over 30 days and three
(2001) Brainstem activation specific to migraine headache. Lan- spontaneous attacks. Brain J Neurol 139(Pt 7):1987–1993
cet 357(9261):1016–1017 45. Peng KP, May A (2019) Migraine understood as a sensory thresh-
old disease. Pain 160(7):1494–1501

13
Journal of Neurology (2023) 270:3654–3666 3661

46. May A, Burstein R (2019) Hypothalamic regulation of headache 66. Granziera C, Daducci A, Romascano D, Roche A, Helms G,
and migraine. Cephalalgia Int J Headache 39(13):1710–1719 Krueger G et al (2014) Structural abnormalities in the thalamus
47. Schulte LH, Mehnert J, May A (2020) Longitudinal neuroim- of migraineurs with aura: a multiparametric study at 3 T. Hum
aging over 30 days: temporal characteristics of migraine. Ann Brain Mapp 35(4):1461–1468
Neurol 87(4):646–651 67. Messina R, Rocca MA, Colombo B, Pagani E, Falini A, Comi G
48. Denuelle M, Fabre N, Payoux P, Chollet F, Geraud G (2007) et al (2015) White matter microstructure abnormalities in pedi-
Hypothalamic activation in spontaneous migraine attacks. Head- atric migraine patients. Cephalalgia 35(14):1278–1286
ache 47(10):1418–1426 68. Hougaard A, Nielsen SH, Gaist D, Puonti O, Garde E, Reislev
49. Maniyar FH, Sprenger T, Monteith T, Schankin C, Goadsby NL et al (2020) Migraine with aura in women is not associ-
PJ (2014) Brain activations in the premonitory phase of nitro- ated with structural thalamic abnormalities. Neuroimage Clin
glycerin-triggered migraine attacks. Brain J Neurol 137(Pt 28:102361
1):232–241 69. Amin FM, Hougaard A, Magon S, Sprenger T, Wolfram F, Ros-
50. Lee MJ, Park BY, Cho S, Kim ST, Park H, Chung CS (2019) trup E et al (2018) Altered thalamic connectivity during spon-
Increased connectivity of pain matrix in chronic migraine: a taneous attacks of migraine without aura: a resting-state fMRI
resting-state functional MRI study. J Headache Pain 20(1):29 study. Cephalalgia 38(7):1237–1244
51. Schulte LH, Allers A, May A (2017) Hypothalamus as a media- 70. Hodkinson DJ, Wilcox SL, Veggeberg R, Noseda R, Burstein R,
tor of chronic migraine: evidence from high-resolution fMRI. Borsook D et al (2016) Increased amplitude of thalamocortical
Neurology 88(21):2011–2016 low-frequency oscillations in patients with migraine. J Neurosci
52. Kagan R, Kainz V, Burstein R, Noseda R (2013) Hypothalamic 36(30):8026–8036
and basal ganglia projections to the posterior thalamus: possible 71. Tu Y, Fu Z, Zeng F, Maleki N, Lan L, Li Z et al (2019) Abnor-
role in modulation of migraine headache and photophobia. Neu- mal thalamocortical network dynamics in migraine. Neurology
roscience 248:359–368 92(23):e2706–e2716
53. Malick A, Burstein R (1998) Cells of origin of the trigeminohy- 72. Lim M, Jassar H, Kim DJ, Nascimento TD, DaSilva AF (2021)
pothalamic tract in the rat. J Comp Neurol 400(1):125–144 Differential alteration of fMRI signal variability in the ascend-
54. Abdallah K, Artola A, Monconduit L, Dallel R, Luccarini P ing trigeminal somatosensory and pain modulatory pathways
(2013) Bilateral descending hypothalamic projections to the in migraine. J Headache Pain 22(1):4
spinal trigeminal nucleus caudalis in rats. PLoS One 8(8):e73022 73. Shields KG, Goadsby PJ (2006) Serotonin receptors modulate
55. Watanabe M, Kopruszinski CM, Moutal A, Ikegami D, Khanna trigeminovascular responses in ventroposteromedial nucleus
R, Chen Y et al (2022) Dysregulation of serum prolactin links of thalamus: a migraine target? Neurobiol Dis 23(3):491–501
the hypothalamus with female nociceptors to promote migraine. 74. Shields KG, Goadsby PJ (2005) Propranolol modulates trigemi-
Brain 145(8):2894–2909 novascular responses in thalamic ventroposteromedial nucleus:
56. Gross EC, Lisicki M, Fischer D, Sandor PS, Schoenen J (2019) a role in migraine? Brain 128(Pt 1):86–97
The metabolic face of migraine—from pathophysiology to treat- 75. Tepe N, Filiz A, Dilekoz E, Akcali D, Sara Y, Charles A et al
ment. Nat Rev Neurol 15(11):627–643 (2015) The thalamic reticular nucleus is activated by cortical
57. Noseda R, Jakubowski M, Kainz V, Borsook D, Burstein R spreading depression in freely moving rats: prevention by acute
(2011) Cortical projections of functionally identified thalamic valproate administration. Eur J Neurosci 41(1):120–128
trigeminovascular neurons: implications for migraine headache 76. Andreou AP, Shields KG, Goadsby PJ (2010) GABA and val-
and its associated symptoms. J Neurosci 31(40):14204–14217 proate modulate trigeminovascular nociceptive transmission
58. Noseda R, Borsook D, Burstein R (2017) Neuropeptides and neu- in the thalamus. Neurobiol Dis 37(2):314–323
rotransmitters that modulate thalamo-cortical pathways relevant 77. Summ O, Charbit AR, Andreou AP, Goadsby PJ (2010) Mod-
to migraine headache. Headache 57(Suppl 2):97–111 ulation of nocioceptive transmission with calcitonin gene-
59. Burstein R, Jakubowski M, Garcia-Nicas E, Kainz V, Bajwa related peptide receptor antagonists in the thalamus. Brain J
Z, Hargreaves R et  al (2010) Thalamic sensitization trans- Neurol 133(9):2540–2548
forms localized pain into widespread allodynia. Ann Neurol 78. Puledda F, Shields K (2018) Non-phar macological
68(1):81–91 approaches for migraine. Neurother J Am Soc Exp Neurother
60. Burstein R, Yamamura H, Malick A, Strassman AM (1998) 15(2):336–345
Chemical stimulation of the intracranial dura induces enhanced 79. Andreou AP, Holland PR, Akerman S, Summ O, Fredrick J,
responses to facial stimulation in brain stem trigeminal neurons. Goadsby PJ (2016) Transcranial magnetic stimulation and poten-
J Neurophysiol 79(2):964–982 tial cortical and trigeminothalamic mechanisms in migraine.
61. Strassman AM, Raymond SA, Burstein R (1996) Sensitization of Brain J Neurol 139(Pt 7):2002–2014
meningeal sensory neurons and the origin of headaches. Nature 80. Olesen J, Larsen B, Lauritzen M (1981) Focal hyperemia fol-
384(6609):560–564 lowed by spreading oligemia and impaired activation of rCBF
62. Suzuki K, Suzuki S, Shiina T, Kobayashi S, Hirata K (2022) in classic migraine. Ann Neurol 9(4):344–352
Central sensitization in migraine: a narrative review. J Pain Res 81. Barral E, Martins Silva E, García-Azorín D, Viana M, Puledda
15:2673–2682 F (2023) Differential diagnosis of visual phenomena associated
63. Noseda R, Bernstein CA, Nir RR, Lee AJ, Fulton AB, Bertisch with migraine: spotlight on aura and visual snow syndrome.
SM et al (2016) Migraine photophobia originating in cone-driven Diagnostics 13(2):252
retinal pathways. Brain 139(Pt 7):1971–1986 82. Leão AAP (1944) Spreading depression of activity in the cerebral
64. Noseda R, Kainz V, Jakubowski M, Gooley JJ, Saper CB, Digre cortex. J Neurophysiol 7:359–390
K et al (2010) A neural mechanism for exacerbation of headache 83. Hadjikhani N, Sanchez Del Rio M, Wu O, Schwartz D, Bakker
by light. Nat Neurosci 13(2):239–245 D, Fischl B et al (2001) Mechanisms of migraine aura revealed
65. Magon S, May A, Stankewitz A, Goadsby PJ, Tso AR, Ashina by functional MRI in human visual cortex. Proc Natl Acad Sci
M et al (2015) Morphological abnormalities of thalamic subnu- USA 98(8):4687–4692
clei in migraine: a multicenter MRI study at 3 Tesla. J Neurosci 84. Bolay H, Reuter U, Dunn AK, Huang Z, Boas DA, Moskowitz
35(40):13800–13806 MA (2002) Intrinsic brain activity triggers trigeminal meningeal
afferents in a migraine model. Nat Med 8(2):136–142

13

3662 Journal of Neurology (2023) 270:3654–3666

85. Zhang X, Levy D, Kainz V, Noseda R, Jakubowski M, Burstein R 105. Dobrynina LA, Suslina AD, Gubanova MV, Belopasova AV,
(2011) Activation of central trigeminovascular neurons by corti- Sergeeva AN, Evers S et al (2021) White matter hyperintensity
cal spreading depression. Ann Neurol 69(5):855–865 in different migraine subtypes. Sci Rep 11(1):10881
86. Melo-Carrillo A, Noseda R, Nir RR, Schain AJ, Stratton J, 106. Ahmed SR, Mohamed AAM, Salem HH, Helmy S, Moustafa RR,
Strassman AM et al (2017) Selective inhibition of trigemino- Borham SMF (2022) Association of white matter hyperintensi-
vascular neurons by fremanezumab: a humanized monoclonal ties with migraine phenotypes and response to treatment. Acta
anti-CGRP antibody. J Neurosci 37(30):7149–7163 Neurol Belg. https://​doi.​org/​10.​1007/​s13760-​022-​02015-x
87. Charles A (2018) The migraine aura. Continuum (Minneapolis, 107. Coppola G, Ambrosini A, Di Clemente L, Magis D, Fumal A,
Minn). 24(4, Headache):1009–1022 Gerard P et al (2007) Interictal abnormalities of gamma band
88. Goadsby PJ (2002) Parallel concept of migraine pathogenesis. activity in visual evoked responses in migraine: an indication
Ann Neurol 51:140 of thalamocortical dysrhythmia? Cephalalgia Int J Headache
89. Messina R, Filippi M, Goadsby PJ (2018) Recent advances in 27(12):1360–1367
headache neuroimaging. Curr Opin Neurol 31(4):379–385 108. Ambrosini A, Rossi P, De Pasqua V, Pierelli F, Schoenen J (2003)
90. Coppola G, Parisi V, Di Renzo A, Pierelli F (2020) Corti- Lack of habituation causes high intensity dependence of auditory
cal pain processing in migraine. J Neural Transm (Vienna) evoked cortical potentials in migraine. Brain J Neurol 126(Pt
127(4):551–566 9):2009–2015
91. Magon S, May A, Stankewitz A, Goadsby PJ, Schankin C, Ashina 109. Afra J, Cecchini AP, De Pasqua V, Albert A, Schoenen J (1998)
M et al (2019) Cortical abnormalities in episodic migraine: a Visual evoked potentials during long periods of pattern-reversal
multi-center 3T MRI study. Cephalalgia 39(5):665–673 stimulation in migraine. Brain J Neurol 121(Pt 2):233–241
92. Park S, Lee DA, Lee HJ, Shin KJ, Park KM (2022) Brain 110. Coppola G, Di Lorenzo C, Schoenen J, Pierelli F (2013) Habit-
networks in migraine with and without aura: an explora- uation and sensitization in primary headaches. J Headache Pain
tory arterial spin labeling MRI study. Acta Neurol Scand 14:65
145(2):208–214 111. Freilinger T, Anttila V, de Vries B, Malik R, Kallela M, Ter-
93. Arngrim N, Hougaard A, Schytz HW, Vestergaard MB, Britze windt GM et al (2012) Genome-wide association analysis iden-
J, Amin FM et al (2019) Effect of hypoxia on BOLD fMRI tifies susceptibility loci for migraine without aura. Nat Genet
response and total cerebral blood flow in migraine with aura 44(7):777–782
patients. J Cereb Blood Flow Metab 39(4):680–689 112. Younis S, Hougaard A, Vestergaard MB, Larsson HBW, Ashina
94. Farago P, Tuka B, Toth E, Szabo N, Kiraly A, Csete G et al M (2017) Migraine and magnetic resonance spectroscopy: a
(2017) Interictal brain activity differs in migraine with and systematic review. Curr Opin Neurol 30(3):246–262
without aura: resting state fMRI study. J Headache Pain 18(1):8 113. Ferrari MD, Goadsby PJ, Burstein R, Kurth T, Ayata C, Charles
95. Vereb D, Szabo N, Tuka B, Tajti J, Kiraly A, Farago P et al A et al (2022) Migraine. Nat Rev Dis Primers 8(1):2
(2020) Temporal instability of salience network activity in 114. Moreno-Ajona D, Villar-Martínez MD, Goadsby PJ (2021) Tar-
migraine with aura. Pain 161(4):856–864 gets for migraine treatment: beyond calcitonin gene-related
96. Russo A, Tessitore A, Silvestro M, Di Nardo F, Trojsi F, Del peptide. Curr Opin Neurol 34(3):363–372
Santo T et al (2019) Advanced visual network and cerebel- 115. Garelja ML, Hay DL (2022) A narrative review of the cal-
lar hyperresponsiveness to trigeminal nociception in migraine citonin peptide family and associated receptors as migraine
with aura. J Headache Pain 20(1):46 targets: calcitonin gene-related peptide and beyond. Headache
97. Puledda F, Ffytche DH, O’Daly O, Goadsby PJ (2019) Imag- 62(9):1093–1104
ing the visual network in the migraine spectrum. Front Neurol 116. Houts CR, McGinley JS, Nishida TK, Buse DC, Wirth RJ,
10:1325 Dodick DW et  al (2021) Systematic review of outcomes
98. Wang ZW, Yin ZH, Wang X, Zhang YT, Xu T, Du JR et al and endpoints in acute migraine clinical trials. Headache
(2022) Brain structural and functional changes during men- 61(2):263–275
strual migraine: relationships with pain. Front Mol Neurosci 117. McGinley JS, Houts CR, Nishida TK, Buse DC, Lipton RB,
15:967103 Goadsby PJ et al (2021) Systematic review of outcomes and
99. Maleki N, Becerra L, Brawn J, Bigal M, Burstein R, Borsook D endpoints in preventive migraine clinical trials. Headache
(2012) Concurrent functional and structural cortical alterations 61(2):253–262
in migraine. Cephalalgia Int J Headache 32(8):607–620 118. Lo SH, Gallop K, Smith T, Powell L, Johnston K, Hubig LT
100. Sheng L, Zhao P, Ma H, Yuan C, Zhong J, Dai Z et al (2020) A et al (2022) Real-world experience of interictal burden and treat-
lack of consistent brain grey matter alterations in migraine. Brain ment in migraine: a qualitative interview study. J Headache Pain
143(6):e45 23(1):65
101. Chen ZH, Cui YL, Sun JT, Li YT, Zhang C, Zhang YM et al 119. Goadsby PJ (2000) The pharmacology of headache. Prog Neu-
(2022) The brain structure and function abnormalities of robiol 62(5):509–525
migraineurs: a systematic review and neuroimaging meta-anal- 120. Goadsby PJ, Edvinsson L (1994) Joint 1994 Wolff Award Presen-
ysis. Front Neurol 13:1022793 tation. Peripheral and central trigeminovascular activation in cat
102. Hamedani AG, Rose KM, Peterlin BL, Mosley TH, Coker LH, is blocked by the serotonin (5HT)-1D receptor agonist 311C90.
Jack CR et al (2013) Migraine and white matter hyperintensities: Headache 34(7):394–399
the ARIC MRI study. Neurology 81(15):1308–1313 121. Ferrari MD, Goadsby PJ, Roon KI, Lipton RB (2002) Triptans
103. Kruit MC, van Buchem MA, Launer LJ, Terwindt GM, Ferrari (serotonin, 5-HT1B/1D agonists) in migraine: detailed results
MD (2010) Migraine is associated with an increased risk of deep and methods of a meta-analysis of 53 trials. Cephalalgia Int J
white matter lesions, subclinical posterior circulation infarcts and Headache 22(8):633–658
brain iron accumulation: the population-based MRI CAMERA 122. Brandes JL, Kudrow D, Stark SR, O’Carroll CP, Adelman JU,
study. Cephalalgia 30(2):129–136 O’Donnell FJ et al (2007) Sumatriptan-naproxen for acute treat-
104. Monteith T, Gardener H, Rundek T, Dong C, Yoshita M, Elkind ment of migraine: a randomized trial. JAMA 297(13):1443–1454
MS et al (2014) Migraine, white matter hyperintensities, and 123. Silberstein SD, Mannix LK, Goldstein J, Couch JR, Byrd SC,
subclinical brain infarction in a diverse community: the northern Ames MH et al (2008) Multimechanistic (sumatriptan-naproxen)
Manhattan study. Stroke 45(6):1830–1832

13
Journal of Neurology (2023) 270:3654–3666 3663

early intervention for the acute treatment of migraine. Neurology 139. Ho TW, Ferrari MD, Dodick DW, Galet V, Kost J, Fan X et al
71(2):114–121 (2008) Efficacy and tolerability of MK-0974 (telcagepant), a
124. van Casteren DS, Kurth T, Danser AHJ, Terwindt GM, Maassen- new oral antagonist of calcitonin gene-related peptide recep-
VanDenBrink A (2021) Sex differences in response to triptans: a tor, compared with zolmitriptan for acute migraine: a ran-
systematic review and meta-analysis. Neurology 96(4):162–170 domised, placebo-controlled, parallel-treatment trial. Lancet
125. Sacco S, Lampl C, Amin FM, Braschinsky M, Deligianni C, 372(9656):2115–2123
Uludüz D et al (2022) European Headache Federation (EHF) 140. Hewitt DJ, Aurora SK, Dodick DW, Goadsby PJ, Ge YJ, Bach-
consensus on the definition of effective treatment of a migraine man R et al (2011) Randomized controlled trial of the CGRP
attack and of triptan failure. J Headache Pain 23(1):133 receptor antagonist MK-3207 in the acute treatment of migraine.
126. Lombard L, Farrar M, Ye W, Kim Y, Cotton S, Buchanan AS Cephalalgia Int J Headache 31(6):712–722
et al (2020) A global real-world assessment of the impact on 141. Olesen J, Diener HC, Husstedt IW, Goadsby PJ, Hall D, Meier U
health-related quality of life and work productivity of migraine in et al (2004) Calcitonin gene-related peptide receptor antagonist
patients with insufficient versus good response to triptan medica- BIBN 4096 BS for the acute treatment of migraine. N Engl J Med
tion. J Headache Pain 21(1):41 350(11):1104–1110
127. Wu JW, Lai PY, Chen YL, Wang YF, Lirng JF, Chen ST et al 142. Diener HC, Barbanti P, Dahlof C, Reuter U, Habeck J, Podhorna
(2022) The use of neuroimaging for predicting sumatriptan treat- J (2011) BI 44370 TA, an oral CGRP antagonist for the treatment
ment response in patients with migraine. Front Neurol 13:798695 of acute migraine attacks: results from a phase II study. Cepha-
128. Roberto G, Raschi E, Piccinni C, Conti V, Vignatelli L, lalgia 31(5):573–584
D’Alessandro R et  al (2015) Adverse cardiovascular events 143. Food and Drug Administration (2020). Drug Approval Package:
associated with triptans and ergotamines for treatment of NURTEC ODT. https://​www.​acces​sdata.​fda.​gov/​drugs​atfda_​
migraine: systematic review of observational studies. Cephalal- docs/​nda/​2020/​21272​8Orig​1s000​TOC.​cfm. Accessed 13 Mar
gia 35(2):118–131 2022
129. Dodick D, Lipton RB, Martin V, Papademetriou V, Rosamond 144. Food and Drug Administration (2019) FDA approves new treat-
W, MaassenVanDenBrink A et al (2004) Consensus statement: ment for adults with migraine. https://w ​ ww.f​ da.g​ ov/n​ ews-e​ vents/​
cardiovascular safety profile of triptans (5-HT agonists) in the press-​annou​nceme​nts/​fda-​appro​ves-​new-​treat​ment-​adults-​migra​
acute treatment of migraine. Headache 44(5):414–425 ine. Accessed 13 Mar 2022
130. Nelson DL, Phebus LA, Johnson KW, Wainscott DB, Cohen ML, 145. Dodick DW, Lipton RB, Ailani J, Lu K, Finnegan M, Trugman
Calligaro DO et al (2010) Preclinical pharmacological profile of JM et al (2019) Ubrogepant for the treatment of migraine. N Engl
the selective 5-HT1F receptor agonist lasmiditan. Cephalalgia J Med 381(23):2230–2241
30(10):1159–1169 146. Lipton RB, Dodick DW, Ailani J, Lu K, Finnegan M, Szegedi
131. Vila-Pueyo M, Page K, Murdock PR, Loraine HJ, Woodrooffe A et al (2019) Effect of ubrogepant vs placebo on pain and the
AJ, Johnson KW et al (2022) The selective 5-HT(1F) receptor most bothersome associated symptom in the acute treatment of
agonist lasmiditan inhibits trigeminal nociceptive processing: migraine: the ACHIEVE II randomized clinical trial. JAMA
implications for migraine and cluster headache. Br J Pharmacol 322(19):1887–1898
179(3):358–370 147. Lipton R, Coric V, Stock E, Stock D, Morris B, McCormack T,
132. Ferrari MD, Färkkilä M, Reuter U, Pilgrim A, Davis C, Krauss et al (2018). Rimegepant 75 mg, an oral calcitonin gene-related
M et al (2010) Acute treatment of migraine with the selective peptide antagonist, for the acute treatment of migraine: two phase
5-HT1F receptor agonist lasmiditan—a randomised proof-of- 3, double-blind, randomized, placebo-controlled trials. Cephala-
concept trial. Cephalalgia Int J Headache 30(10):1170–1178 lgia Int J Headache
133. Färkkilä M, Diener H-C, Géraud G, Láinez M, Schoenen J, 148. Lipton RB, Croop R, Stock EG, Stock DA, Morris BA, Frost M
Harner N et al (2012) Efficacy and tolerability of lasmiditan, et al (2019) Rimegepant, an oral calcitonin gene-related peptide
an oral 5-HT1F receptor agonist, for the acute treatment of receptor antagonist, for migraine. N Engl J Med 381(2):142–149
migraine: a phase 2 randomised, placebo-controlled, parallel- 149. Croop R, Goadsby PJ, Stock DA, Conway CM, Forshaw M, Stock
group, dose-ranging study. Lancet Neurol 11(5):405–413 EG et al (2019) Efficacy, safety, and tolerability of rimegepant
134. Kuca B, Silberstein SD, Wietecha L, Berg PH, Dozier G, orally disintegrating tablet for the acute treatment of migraine:
Lipton RB (2018) Lasmiditan is an effective acute treat- a randomised, phase 3, double-blind, placebo-controlled trial.
ment for migraine: a phase 3 randomized study. Neurology Lancet 394(10200):737–745
91(24):e2222–e2232 150. Croop R, Madonia J, Stock DA, Thiry A, Forshaw M, Murphy
135. Goadsby PJ, Wietecha LA, Dennehy EB, Kuca B, Case MG, A et al (2022) Zavegepant nasal spray for the acute treatment
Aurora SK et al (2019) Phase 3 randomized, placebo-controlled, of migraine: a Phase 2/3 double-blind, randomized, placebo-
double-blind study of lasmiditan for acute treatment of migraine. controlled, dose-ranging trial. Headache 62(9):1153–1163
Brain J Neurol 142(7):1894–1904 151. Moreno-Ajona D, Villar-Martinez MD, Goadsby PJ (2022) New
136. Ashina M, Reuter U, Smith T, Krikke-Workel J, Klise SR, Bragg generation gepants: migraine acute and preventive medications.
S et al (2021) Randomized, controlled trial of lasmiditan over J Clin Med 11(6):1656
four migraine attacks: findings from the CENTURION study. 152. Hutchinson S, Dodick DW, Treppendahl C, Bennett NL, Yu
Cephalalgia Int J Headache 41(3):294–304 SY, Guo H et  al (2021) Ubrogepant for the acute treatment
137. Shapiro RE, Hochstetler HM, Dennehy EB, Khanna R, Doty EG, of migraine: pooled efficacy, safety, and tolerability from the
Berg PH et al (2019) Lasmiditan for acute treatment of migraine ACHIEVE I and ACHIEVE II phase 3 randomized trials. Neurol
in patients with cardiovascular risk factors: post-hoc analysis Ther 10(1):235–249
of pooled results from 2 randomized, double-blind, placebo- 153. Al-Hassany L, Goadsby PJ, Danser AHJ, MaassenVanDenBrink
controlled, phase 3 trials. J Headache Pain 20(1):90 A (2022) Calcitonin gene-related peptide-targeting drugs for
138. Puledda F, Younis S, Huessler E-M, Haghdoost F, Lisicki M, migraine: how pharmacology might inform treatment decisions.
Goadsby PJ et al (2023) Efficacy, safety and indirect comparisons Lancet Neurol 21(3):284–294
of lasmiditan, rimegepant, and ubrogepant for the acute treatment 154. Sang CN, Ramadan NM, Wallihan RG, Chappell AS, Freitag
of migraine: a systematic review and network meta-analysis of FG, Smith TR et al (2004) LY293558, a novel AMPA/GluR5
the literature. Cephalalgia Int J Headache 43(3):0333

13

3664 Journal of Neurology (2023) 270:3654–3666

antagonist, is efficacious and well-tolerated in acute migraine. double-blind, placebo-controlled study (PROMISE-1). Cephala-
Cephalalgia 24(7):596–602 lgia Int J Headache 40(3):241–254
155. Waung MW, Akerman S, Wakefield M, Keywood C, Goadsby PJ 172. Lipton RB, Goadsby PJ, Smith J, Schaeffler BA, Biondi DM,
(2016) Metabotropic glutamate receptor 5: a target for migraine Hirman J et  al (2020) Efficacy and safety of eptinezumab
therapy. Ann Clin Transl Neurol 3(8):560–571 in patients with chronic migraine: PROMISE-2. Neurology
156. Gomez-Mancilla B, Brand R, Jurgens TP, Gobel H, Sommer C, 94(13):e1365–e1377
Straube A et al (2014) Randomized, multicenter trial to assess the 173. Mulleners WM, Kim B-K, Láinez MJA, Lanteri-Minet M, Pozo-
efficacy, safety and tolerability of a single dose of a novel AMPA Rosich P, Wang S et al (2020) Safety and efficacy of galcan-
receptor antagonist BGG492 for the treatment of acute migraine ezumab in patients for whom previous migraine preventive medi-
attacks. Cephalalgia 34(2):103–113 cation from two to four categories had failed (CONQUER): a
157. Goadsby PJ (2013) Therapeutic prospects for migraine: can para- multicentre, randomised, double-blind, placebo-controlled, phase
dise be regained? Ann Neurol 74(3):423–434 3b trial. Lancet Neurol 19(10):814–825
158. Afridi SK, Giffin NJ, Kaube H, Goadsby PJ (2013) A randomized 174. Ashina M, Lanteri-Minet M, Pozo-Rosich P, Ettrup A, Christ-
controlled trial of intranasal ketamine in migraine with prolonged offersen CL, Josiassen MK et al (2022) Safety and efficacy of
aura. Neurology 80(7):642–647 eptinezumab for migraine prevention in patients with two-to-four
159. Eigenbrodt AK, Ashina H, Khan S, Diener H-C, Mitsikostas DD, previous preventive treatment failures (DELIVER): a multi-arm,
Sinclair AJ et al (2021) Diagnosis and management of migraine randomised, double-blind, placebo-controlled, phase 3b trial.
in ten steps. Nat Rev Neurol 17(8):501–514 Lancet Neurol 21(7):597–607
160. Diener HC, Charles A, Goadsby PJ, Holle D (2015) New thera- 175. Haghdoost F, Puledda F, Huessler EM, Messina R, Pozo-Rosich
peutic approaches for the prevention and treatment of migraine. P (2023) Evaluating the efficacy of CGRP mAbs and gepants for
Lancet Neurol 14(10):1010–1022 the preventive treatment of migraine: a systematic review and
161. American HS (2019) The American Headache Society position network meta-analysis of phase 3 randomised controlled trials.
statement on integrating new migraine treatments into clinical Cephalalgia Int J Headache 43(3):0331
practice. Headache 59(1):1–18 176. Messina R, Huessler EM, Puledda F, Haghdoost F, Lebedeva
162. Goadsby PJ, Reuter U, Hallström Y, Broessner G, Bonner JH, ER, Diener HC (2023) Safety and tolerability of monoclonal
Zhang F et al (2017) A controlled trial of erenumab for epi- antibodies targeting the CGRP pathway and gepants in migraine
sodic migraine. N Engl J Med 377(22):2123–2132 prevention: a systematic review and meta-analysis. Cephalalgia
163. Reuter U, Goadsby PJ, Lanteri-Minet M, Wen S, Hours- Int J Headache 43(3):0331
Zesiger P, Ferrari MD et al (2018) Efficacy and tolerability 177. Vernieri F, Brunelli N, Messina R, Costa CM, Colombo B,
of erenumab in patients with episodic migraine in whom two- Torelli P et al (2021) Discontinuing monoclonal antibodies tar-
to-four previous preventive treatments were unsuccessful: a geting CGRP pathway after one-year treatment: an observational
randomised, double-blind, placebo-controlled, phase 3b study. longitudinal cohort study. J Headache Pain 22(1):154
Lancet 392(10161):2280–2287 178. Nsaka M, Scheffler A, Wurthmann S, Schenk H, Kleinschnitz
164. Dodick DW, Ashina M, Brandes JL, Kudrow D, Lanteri-Minet C, Glas M et al (2022) Real-world evidence following a manda-
M, Osipova V et al (2018) ARISE: a phase 3 randomized trial tory treatment break after a 1-year prophylactic treatment with
of erenumab for episodic migraine. Cephalalgia Int J Headache calcitonin gene-related peptide (pathway) monoclonal antibodies.
38(6):1026–1037 Brain Behav 12(7):e2662
165. Silberstein SD, Dodick DW, Bigal ME, Yeung PP, Goad- 179. di Cola FS, Caratozzolo S, Venturelli E, Balducci U, Sidoti V,
sby PJ, Blankenbiller T et al (2017) Fremanezumab for the Pari E et al (2021) Erenumab discontinuation after 12-month
preventive treatment of chronic migraine. N Engl J Med treatment: a multicentric, observational real-life study. Neurol
377(22):2113–2122 Clin Pract 11(6):e834–e900
166. Ferrari MD, Diener HC, Ning X, Galic M, Cohen JM, Yang R 180. Terhart M, Mecklenburg J, Neeb L, Overeem LH, Siebert A,
et al (2019) Fremanezumab versus placebo for migraine preven- Steinicke M et al (2021) Deterioration of headache impact and
tion in patients with documented failure to up to four migraine health-related quality of life in migraine patients after cessa-
preventive medication classes (FOCUS): a randomised, double- tion of preventive treatment with CGRP(-receptor) antibodies.
blind, placebo-controlled, phase 3b trial. Lancet (London, Eng- J Headache Pain 22(1):158
land) 394(10203):1030–1040 181. Sacco S, Amin FM, Ashina M, Bendtsen L, Deligianni CI, Gil-
167. Dodick DW, Silberstein SD, Bigal ME, Yeung PP, Goadsby PJ, Gouveia R et al (2022) European Headache Federation guideline
Blankenbiller T et al (2018) Effect of fremanezumab compared on the use of monoclonal antibodies targeting the calcitonin gene
with placebo for prevention of episodic migraine: a randomized related peptide pathway for migraine prevention—2022 update.
clinical trial. JAMA 319(19):1999–2008 J Headache Pain 23(1):67
168. Detke HC, Goadsby PJ, Wang S, Friedman DI, Selzler KJ, Aurora 182. Ailani J, Lipton RB, Goadsby PJ, Guo H, Miceli R, Severt L
SK (2018) Galcanezumab in chronic migraine. The randomized, et al (2021) Atogepant for the preventive treatment of migraine.
double-blind, placebo-controlled REGAIN study. Neurology N Engl J Med 385(8):695–706
91(24):e2211–e2221 183. Goadsby PJ, Dodick DW, Ailani J, Trugman JM, Finnegan M,
169. Stauffer VL, Dodick DW, Zhang Q, Carter JN, Ailani J, Conley Lu K et  al (2020) Safety, tolerability, and efficacy of orally
RR (2018) Evaluation of galcanezumab for the prevention of epi- administered atogepant for the prevention of episodic migraine
sodic migraine: the EVOLVE-1 randomized clinical trial. JAMA in adults: a double-blind, randomised phase 2b/3 trial. Lancet
Neurol 75(9):1080–1088 Neurol 19(9):727–737
170. Skljarevski V, Matharu M, Millen BA, Ossipov MH, Kim BK, 184. Croop R, Lipton RB, Kudrow D, Stock DA, Kamen L, Conway
Yang JY (2018) Efficacy and safety of galcanezumab for the pre- CM et al (2021) Oral rimegepant for preventive treatment of
vention of episodic migraine: results of the EVOLVE-2 Phase 3 migraine: a phase 2/3, randomised, double-blind, placebo-con-
randomized controlled clinical trial. Cephalalgia Int J Headache trolled trial. Lancet (London, England) 397(10268):51–60
38(8):1442–1454 185. Boinpally R, Jakate A, Butler M, Borbridge L, Periclou A (2021)
171. Ashina M, Saper J, Cady R, Schaeffler BA, Biondi DM, Hirman Single-dose pharmacokinetics and safety of atogepant in adults
J et al (2020) Eptinezumab in episodic migraine: a randomized,

13
Journal of Neurology (2023) 270:3654–3666 3665

with hepatic impairment: results from an open-label, phase 1 201. Zhou T, Tang Y, Zhu H (2022) Effectiveness and safety of
trial. Clin Pharmacol Drug Dev 10(7):726–733 memantine for headache: a meta-analysis of randomized con-
186. Tong G, Savant I, Jariwala N, Burt D, Zheng N, Buzescu A et al trolled studies. Clin Neuropharmacol 45(3):40–44
(2013) Phase I single and multiple dose study to evaluate the 202. Puledda F, Goadsby PJ (2017) An update on non-pharmacolog-
safety, tolerability. J Headache Pain 14(Suppl 1):118 ical neuromodulation for the acute and preventive treatment of
187. AbbVie. (2021) QULIPTA (atogepant) tablets, for oral use: US migraine. Headache 57(4):685–691
prescribing information. https://​www.​acces​sdata.​fda.​gov/​drugs​ 203. Lipton RB, Dodick DW, Silberstein SD, Saper JR, Aurora SK,
atfda_​docs/​label/​2021/​21520​6Orig​1s000​lbl.​pdf. Accessed 13 Pearlman SH et al (2010) Single-pulse transcranial magnetic
Mar 2022 stimulation for acute treatment of migraine with aura: a ran-
188. Min KC, Kraft WK, Bondiskey P, Colón-González F, Liu W, Xu domised, double-blind, parallel-group, sham-controlled trial.
J et al (2021) Atogepant is not associated with clinically mean- Lancet Neurol 9(4):373–380
ingful alanine aminotransferase elevations in healthy adults. Clin 204. Bhola R, Kinsella E, Giffin N, Lipscombe S, Ahmed F, Weath-
Transl Sci 14(2):599–605 erall M et al (2015) Single-pulse transcranial magnetic stimula-
189. Boinpally R, McNamee B, Yao L, Butler M, McGeeney D, Bor- tion (sTMS) for the acute treatment of migraine: evaluation of
bridge L et al (2021) A single supratherapeutic dose of atogepant outcome data for the UK post market pilot program. J Headache
does not affect cardiac repolarization in healthy adults: results Pain 16:535
from a randomized, single-dose, phase 1 crossover trial. Clin 205. Starling AJ, Tepper SJ, Marmura MJ, Shamim EA, Robbins MS,
Pharmacol Drug Dev 10(9):1099–1107 Hindiyeh N et al (2018) A multicenter, prospective, single arm,
190. Pozo-Rosich P, Ailani J, Ashina M, Goadsby PJ, Lipton R, open label, observational study of sTMS for migraine prevention
Reuter U et al (2022) Atogepant for the preventive treatment (ESPOUSE Study). Cephalalgia Int J Head 38(6):1038–1048
of chronic migraine: results from the PROGRESS phase 3 trial. 206. Lloyd JO, Hill B, Murphy M, Al-Kaisy A, Andreou AP, Lambru
Cephalalgia Int J Headache 42(1S):14–15 G (2022) Single-pulse transcranial magnetic stimulation for the
191. Lanteri-Minet M, Ducros A, Francois C, Olewinska E, Niko- preventive treatment of difficult-to-treat migraine: a 12-month
dem M, Dupont-Benjamin L (2022) Effectiveness of onabotu- prospective analysis. J Headache Pain 23(1):63
linumtoxinA (BOTOX(R)) for the preventive treatment of 207. Chou DE, Shnayderman Yugrakh M, Winegarner D, Rowe V,
chronic migraine: a meta-analysis on 10 years of real-world Kuruvilla D, Schoenen J (2019) Acute migraine therapy with
data. Cephalalgia 42(14):1543–1564 external trigeminal neurostimulation (ACME): a randomized
192. Armanious M, Khalil N, Lu Y, Jimenez-Sanders R (2021) Ere- controlled trial. Cephalalgia Int J Headache 39(1):3–14
numab and OnabotulinumtoxinA combination therapy for the 208. Schoenen J, Vandersmissen B, Jeangette S, Herroelen L, Van-
prevention of intractable chronic migraine without aura: a ret- denheede M, Gerard P et al (2013) Migraine prevention with a
rospective analysis. J Pain Palliat Care Pharmacother 35(1):1–6 supraorbital transcutaneous stimulator: a randomized controlled
193. Scuteri D, Tonin P, Nicotera P, Vulnera M, Altieri GC, Tar- trial. Neurology 80(8):697–704
sitano A et al (2022) Pooled analysis of real-world evidence 209. Vikelis M, Dermitzakis EV, Spingos KC, Vasiliadis GG, Vlachos
supports anti-CGRP mAbs and OnabotulinumtoxinA combined GS, Kararizou E (2017) Clinical experience with transcutane-
trial in chronic migraine. Toxins (Basel). 14(8):529 ous supraorbital nerve stimulation in patients with refractory
194. Cohen F, Armand C, Lipton RB, Vollbracht S (2021) Effi- migraine or with migraine and intolerance to topiramate: a pro-
cacy and tolerability of calcitonin gene-related peptide-tar- spective exploratory clinical study. BMC Neurol 17(1):97
geted monoclonal antibody medications as add-on therapy to 210. Ordas CM, Cuadrado ML, Pareja JA, de-Las-Casas-Camara
OnabotulinumtoxinA in patients with chronic migraine. Pain G, Gomez-Vicente L, Torres-Gaona G et al (2020) Transcu-
Med 22(8):1857–1863 taneous supraorbital stimulation as a preventive treatment for
195. Chowdhury D, Bansal L, Duggal A, Datta D, Mundra A, chronic migraine: a prospective, open-label study. Pain Med
Krishnan A et al (2022) TOP-PRO study: a randomized dou- 21(2):415–422
ble-blind controlled trial of topiramate versus propranolol for 211. Tassorelli C, Grazzi L, de Tommaso M, Pierangeli G, Martelletti
prevention of chronic migraine. Cephalalgia 42(4–5):396–408 P, Rainero I et al (2018) Noninvasive vagus nerve stimulation
196. Lai KL, Niddam DM, Fuh JL, Chen SP, Wang YF, Chen as acute therapy for migraine: the randomized PRESTO study.
WT et al (2017) Flunarizine versus topiramate for chronic Neurology 91(4):e364–e373
migraine prophylaxis: a randomized trial. Acta Neurol Scand 212. Diener HC, Goadsby PJ, Ashina M, Al-Karagholi MA, Sinclair
135(4):476–483 A, Mitsikostas D et al (2019) Non-invasive vagus nerve stimula-
197. Messina R, Lastarria Perez CP, Filippi M, Goadsby PJ (2020) tion (nVNS) for the preventive treatment of episodic migraine:
Candesartan in migraine prevention: results from a retrospective the multicentre, double-blind, randomised, sham-controlled
real-world study. J Neurol 267(11):3243–3247 PREMIUM trial. Cephalalgia 39(12):1475–1487
198. Sanchez-Rodriguez C, Sierra A, Planchuelo-Gomez A, Mar- 213. Silberstein SD, Calhoun AH, Lipton RB, Grosberg BM, Cady
tinez-Pias E, Guerrero AL, Garcia-Azorin D (2021) Real world RK, Dorlas S et al (2016) Chronic migraine headache prevention
effectiveness and tolerability of candesartan in the treatment of with noninvasive vagus nerve stimulation: the EVENT study.
migraine: a retrospective cohort study. Sci Rep 11(1):3846 Neurology 87(5):529–538
199. Liampas I, Siokas V, Brotis A, Vikelis M, Dardiotis E (2020) 214. Najib U, Smith T, Hindiyeh N, Saper J, Nye B, Ashina S et al
Endogenous melatonin levels and therapeutic use of exogenous (2022) Non-invasive vagus nerve stimulation for prevention of
melatonin in migraine: systematic review and meta-analysis. migraine: the multicenter, randomized, double-blind, sham-
Headache 60(7):1273–1299 controlled PREMIUM II trial. Cephalalgia Int J Headache
200. Tseng PT, Yang CP, Su KP, Chen TY, Wu YC, Tu YK et al (2020) 42(7):560–569
The association between melatonin and episodic migraine: a pilot 215. Chen SP, Ay I, de Morais AL, Qin T, Zheng Y, Sadeghian H
network meta-analysis of randomized controlled trials to com- et al (2016) Vagus nerve stimulation inhibits cortical spreading
pare the prophylactic effects with exogenous melatonin supple- depression. Pain 157(4):797–805
mentation and pharmacotherapy. J Pineal Res 69(2):e12663 216. Akerman S, Simon B, Romero-Reyes M (2017) Vagus nerve stim-
ulation suppresses acute noxious activation of trigeminocervical

13

3666 Journal of Neurology (2023) 270:3654–3666

neurons in animal models of primary headache. Neurobiol Dis 222. Nierenburg H, Vieira JR, Lev N, Lin T, Harris D, Vizel M et al
102:96–104 (2020) Remote electrical neuromodulation for the acute treat-
217. Antal A, Kriener N, Lang N, Boros K, Paulus W (2011) Cathodal ment of migraine in patients with chronic migraine: an open-
transcranial direct current stimulation of the visual cortex in the label pilot study. Pain Ther 9(2):531–543
prophylactic treatment of migraine. Cephalalgia Int J Headache 223. Renner T, Sollmann N, Trepte-Freisleder F, Albers L, Mathonia
31(7):820–828 NM, Bonfert MV et al (2019) Repetitive peripheral magnetic
218. Vigano A, D’Elia TS, Sava SL, Auve M, De Pasqua V, Colosimo stimulation (rPMS) in subjects with migraine-setup presentation
A et al (2013) Transcranial direct current stimulation (tDCS) of and effects on skeletal musculature. Front Neurol 10:738
the visual cortex: a proof-of-concept study based on interictal 224. Borner C, Renner T, Trepte-Freisleder F, Urban G, Schandel-
electrophysiological abnormalities in migraine. J Headache Pain maier P, Lang M et al (2022) Response predictors of repetitive
14:23 neuromuscular magnetic stimulation in the preventive treatment
219. Viganò A, Toscano M, Puledda F, Di Piero V (2019) Treating of episodic migraine. Front Neurol 13:919623
chronic migraine with neuromodulation: the role of neurophysi- 225. Tepper SJ, Grosberg B, Daniel O, Kuruvilla DE, Vainstein G,
ological abnormalities and maladaptive plasticity. Front Pharma- Deutsch L et al (2022) Migraine treatment with external con-
col 10:32 current occipital and trigeminal neurostimulation-A randomized
220. Nir RR, Yarnitsky D (2015) Conditioned pain modulation. Curr controlled trial. Headache 62(8):989–1001
Opin Support Palliat Care 9(2):131–137 226. Daniel O, Tepper SJ, Deutsch L, Sharon R (2022) External
221. Yarnitsky D, Dodick DW, Grosberg BM, Burstein R, Ironi A, concurrent occipital and trigeminal neurostimulation relieves
Harris D et al (2019) Remote electrical neuromodulation (REN) migraine headache: a prospective, randomized, double-blind,
relieves acute migraine: a randomized, double-blind, placebo- Sham-controlled trial. Pain Ther 11(3):907–922
controlled, multicenter trial. Headache 59(8):1240–1252

13

You might also like