You are on page 1of 15

Review

Cephalalgia
2020, Vol. 40(1) 107–121
Vestibular migraine: An update on current ! International Headache Society 2019
Article reuse guidelines:

understanding and future directions sagepub.com/journals-permissions


DOI: 10.1177/0333102419869317
journals.sagepub.com/home/cep

Tzu-Chou Huang1,2 , Shuu-Jiun Wang3,4 and


Amir Kheradmand1,5

Abstract
Background: Vestibular migraine is among the most common causes of recurrent vertigo in the general population.
Despite its prevalence and high impact on healthcare cost and utilization, it has remained an under-recognized condition
with largely unknown pathophysiology. In the present article, we aim to provide an overview of the current understand-
ing of vestibular migraine.
Methods: We undertook a narrative literature review on the epidemiology, presentations, clinical and laboratory
findings, pathophysiology, and treatments of vestibular migraine.
Results: Currently, the diagnosis of vestibular migraine relies solely on clinical symptoms since clinical tests of vestibular
function are typically normal, or difficult to interpret based on inconsistent results reported in earlier studies. The
challenges related to diagnosis of vestibular migraine lie in its relatively broad spectrum of manifestations, the absence of
typical migraine headaches with vestibular symptoms, and its very recent definition as a distinct entity. Here, we highlight
these challenges, discuss common vestibular symptoms and clinical presentations in vestibular migraine, and review the
current aspects of its clinical diagnosis and evaluation. The concepts related to the pathophysiology and treatment of
vestibular migraine are also discussed.
Conclusion: Vestibular migraine is still underdiagnosed clinically. Future studies are needed to address the pathophysio-
logical mechanisms and investigate effective treatment regimens.

Keywords
Vestibular migraine, vestibular, migraine, dizziness, vertigo, headache
Date received: 18 March 2019; revised: 30 June 2019; accepted: 7 July 2019

Introduction
Clinicians are no strangers to patients with migraine between the vestibular symptoms and migraine is also
symptoms and frequent dizziness. As a common neuro- reflected by a higher incidence of migraine in patients
logical disorder, migraine affects about 15% of the with recurrent dizziness who do not fulfill the criteria
general population (1). Dizziness is also a common for other vestibular disorders (18–23). Such clinical
symptom, accounting for up to 15% of visits in front-
line healthcare settings (2,3). Considering the preva-
1
lence of both conditions, an overlap in the clinical Department of Neurology, The Johns Hopkins University School of
presentations of vestibular symptoms and migraine Medicine, Baltimore, MD, USA
2
Living Water Neurological Clinic, Tainan, Taiwan
may not seem surprising at first. Several studies, how- 3
Neurological Institute, Taipei-Veterans General Hospital, Taipei, Taiwan
ever, have found a close association between dizziness 4
Brain Research Center and School of Medicine, National Yang-Ming
and migraine beyond coincidence (4–15). Dizziness is University, Taipei, Taiwan
5
more common in migraineurs compared with those suf- Department of Otolaryngology-Head and Neck Surgery, The Johns
fering from other headache subtypes such as tension- Hopkins University School of Medicine, Baltimore, MD, USA
type headache, suggesting a pathological link between
Corresponding author:
migraine and dizziness (9,16,17). In some migraine Shuu-Jiun Wang, Neurological Institute, Taipei Veterans General Hospital,
patients, dizziness or vertigo is even more prominent #201, Shi-Pai Road, Section 2, Taipei 112, Taiwan.
and debilitating than the headache (12). The link Email: sjwang@vghtpe.gov.tw
108 Cephalalgia 40(1)

findings have prompted efforts to recognize and classify Patients with VM symptoms frequently report sen-
migraine-associated dizziness under a distinct diagnos- sitivity to head motion and visual surroundings or dis-
tic entity. abling misperceptions such as a sudden feeling of
Historically, different criteria have been applied to imbalance or tilt. When it comes to characterizing ves-
define a clinical entity that can incorporate vestibular tibular symptoms, dizziness or vertigo are often loosely
and migraine symptoms and, along the way, various used terms that may convey different symptom experi-
terms such as migraine-associated dizziness, migraine- ences in different people. According to the ICVD (28),
related vestibulopathy, migrainous vertigo, benign vertigo is a sensation of self-motion when no self-
recurrent vertigo, and vestibular migraine have been motion is occurring or a sensation of distorted self-
proposed (5,7,8,11–13,15). Among these terms, vestibu- motion during an otherwise normal head movement.
lar migraine (VM) has become widely accepted as it Dizziness is defined as a sensation of impaired spatial
links vestibular and migraine symptoms, and it is also orientation without a distorted sense of motion. In fact,
compatible with the terminology of the International dizziness is a broader term that encompasses false spin-
Classification of Headache Disorders (ICHD). The ning sensations (i.e. spinning vertigo) and also other
operational criteria for VM diagnosis were proposed false sensations such as swaying, tilting, or veering
in 2004 by Neuhauser et al. (24). These criteria were (i.e. non-spinning vertigo). In this classification,
later validated and revised with the consensus of the having dizziness does not preempt inclusion of vertigo
International Headache Society (IHS) and the commit- if both symptoms are present as defined by their cri-
tee for the International Classification of Vestibular teria. Vestibular symptoms do not include a pure sen-
Disorders (ICVD) of the Bárány Society (Table 1) sation of impending fainting (presyncope), disordered
(25–27). As included in the VM diagnostic criteria, thinking (mental confusion), or detachment from real-
the presence or history of migraine is essential for its ity (depersonalization). Likewise, the term dizziness
diagnosis, but the headache and vestibular symptoms should not be applied if the patient experiences a gen-
do not need to temporally coincide. eralized weakness or a non-specific sense of malaise or
fatigue.
Unlike a typical migraine aura, which can last
Table 1. Diagnostic criteria for vestibular migraine based on 5–60 minutes, vestibular symptoms in VM sufferers
the International Classification of Headache Disorders, 3rd can persist for hours, leading to severe impairment of
edition (ICHD-3) (25) and the International Classification of daily activities (25,26,29,30). This is a common clinical
Vestibular Disorders (ICVD) of the Bárány Society (26). scenario in VM patients, which often becomes challen-
ging for clinicians and costly for patients to treat, indi-
Vestibular migraine (ICHD-3 and ICVD) cating an unmet need to delineate VM pathophysiology
A. At least five episodes fulfilling criteria C and D
(31). In this article, we review common VM symptoms
B. A current or past history of 1.1 Migraine without aura or 1.2
and clinical presentations. The current concepts related
Migraine with aura
C. Vestibular symptoms of moderate or severe intensity, to VM pathophysiology and its treatment are dis-
lasting between 5 minutes and 72 hours cussed. Also, some of the barriers in addressing the
D. At least 50% of episodes are associated with at least one of neural mechanisms of dizziness, vertigo, and visuo-
the following three migrainous features: spatial symptoms in VM patients are highlighted.
1. Headache with at least two of the following four charac-
teristics:
a) Unilateral location Epidemiology and demographic factors
b) Pulsating quality Vestibular migraine is a common cause of spontaneous
c) Moderate or severe intensity vertigo in both children and adults (32–34). In children,
d) Aggravation by routine physical activity
benign paroxysmal vertigo of childhood is considered
2. Photophobia and phonophobia
an early manifestation of migraine affecting about 3%
3. Visual aura
E. Not better accounted for by another ICHD-3 diagnosis or of children between ages 6 and 12 (35,36). A recent
by another vestibular disorder nationwide study in the United States based on the
Probable vestibular migraine (ICVD) ICHD-3 criteria found a VM prevalence of 2.7% in
A. At least five episodes with vestibular symptoms of moder- adults (37). In a large population study in Germany,
ate or severe intensity, lasting 5 min to 72 hours VM lifetime prevalence was estimated at about 1%,
B. Only one of the criteria B and D for vestibular migraine is and its one-year prevalence was about 1% (12). In a
fulfilled (migraine history or migraine features during the community-based study of middle-aged women, the
episode) one-year VM prevalence was higher, at about
C. Not better accounted for another vestibular or ICHD 5% (38). Despite classification efforts in recent years,
diagnosis
VM has remained clinically underdiagnosed.
Huang et al. 109

Vestibular migraine is reported to account for 4–10% headache subsides, and in some patients headache
of diagnoses in specialized dizziness and headache and vestibular symptoms never occur together. Also,
clinics, although these numbers reflect different inclu- a minority of VM patients may experience vestibular
sion criteria among various studies (11,39–41). symptoms in the time frame of 5–60 minutes, as defined
According to a study that examined data from a ter- for the duration of aura, and even fewer patients have
tiary dizziness center, only 2% of patients were sus- these symptoms immediately before the headache
pected to have VM by referring doctors, whereas starts. Apart from the headache and vestibular symp-
20% were later diagnosed as VM by specialists (42). toms, VM patients may experience photophobia, pho-
In a Korean multicenter study, about 10% of migrain- nophobia, or visual aura. In a case series, 60% of VM
eurs were diagnosed with VM in their first visit to neur- patients reported phonophobia, 70% photophobia, and
ology clinics (43). Another study found that only 10% 36% migraine auras (11). In another study, 87% of
of patients who met the VM diagnostic criteria were VM patients had photophobia, 86% phonophobia,
told that migraine was the cause of their vestibular 64% headache, and 13% migraine auras (52).
symptoms (37). Phonophobia refers to a sound-induced discomfort,
Similar to other subtypes of migraine, VM has a which is a bilateral, transient phenomenon distinct
female preponderance, with a reported female to male from unilateral or persistent aural symptoms. Other
ratio of 1.5–5 to 1 (5,7,8,11,12,44,45). The reported age intermittent auditory symptoms, such as a sense of
of onset for VM symptoms is between 8 and 50 years pressure or fullness in the ears or tinnitus, have been
old or even older (median ages being the mid-30s reported in up to 40% of VM patients (5,8,9,52–55).
to 40s) (12,21,39). Usually, migraine headache tends However, since these symptoms are non-specific and
to present first, and patients may be headache-free also common in other vestibular disorders, they were
for years before the onset of vestibular symptoms. In not included in the VM diagnostic criteria (26). Of note,
one study, the mean duration between the onset of migraine features must be present at least half of the
headache and vestibular symptoms was about eight time along with vestibular symptoms in order to fulfill
years (46). In women, vestibular symptoms can the criteria for VM diagnosis. At least one migraine
become more pronounced around the time of meno- feature is needed along with the vestibular symptoms,
pause (47). but different migraine features may be present at differ-
In keeping with the heritability and genetic back- ent times. Although the co-occurrence of migraine and
ground of migraine disorders, familial occurrence has vestibular symptoms is required by the ICHD-3 criteria
been reported in some VM patients, with an autosomal for VM diagnosis, the Bárány classification ICVD
dominant pattern of inheritance and a decreased pene- has recognized the heterogeneity of migraine presen-
trance in men (48). Within a family, there could be tation in VM patients, and those patients who only
multiple affected individuals, some with VM and have a history of migraine without co-occurrence of
some with other migraine variants. In a survey of migraine features and vestibular symptoms are included
family members in patients with chronic vertigo under a subcategory of probable vestibular migraine
(and no auditory or neurologic disorders), half of the (Table 1) (26).
biological relatives who reported vertigo met the Vestibular symptoms: In VM patients, vestibular
diagnostic criteria for migraine, as opposed to a small symptoms are generally triggered or aggravated by
percentage of unrelated spouses who also reported changing position, self-motion, or visual motion
vertigo (13). within the surrounding environment. These patients
may describe their symptoms as a sense of spinning,
Clinical presentation: Basis for VM floating, rocking, tilting, swaying, or as being off-
balance, off-kilter, lightheaded, foggy, or unsteady
diagnostic criteria
(Table 2). These various terms may reflect different
Common VM features: Vestibular migraine patients internally experienced phenomena. Analogies such as
may have a history of migraine earlier on in their feeling drunk or sea-sick, getting off a roller-coaster
lives, and their migraine symptoms are often variable or merry-go-round, or even walking on air or pillows
in nature, duration, or temporal relation to their ves- are also commonly used by the patients. Such diverse
tibular symptoms (5,11,49). The core migraine features nature of vestibular symptoms is recognized in the VM
in VM patients are similar to those of migraine with or diagnostic criteria as different forms of vertigo or
without aura. VM patients also report similar migraine dizziness, including spontaneous vertigo, positional
triggers such as sleep disruption, menstruation, stress, vertigo, visually induced vertigo, and head motion-
or specific foods (e.g. aged cheese, red wine, or mono- induced vertigo or dizziness (Table 2). Spontaneous
sodium glutamate) (11,50,51). Not uncommonly, VM vertigo is defined as a false sense of motion, either
patients develop vestibular symptoms after the as internal vertigo with primarily a sensation of
110 Cephalalgia 40(1)

Table 2. Vestibular symptoms in vestibular migraine patients these symptoms are non-specific and may also
and relevant terminology defined by the International occur with other vestibular disorders (9,10,60–62).
Classification of Vestibular Disorders (ICVD). Vestibular symptoms are considered moderate when
Terms usually used by patients
they interfere with daily activities and considered
Spinning, rocking, tilting or swaying severe when they prevent daily activities.
Unsteady, off-balance or off-kilter Clinical overlap in VM presentations: Any vestibular
Lightheaded disorder can be complicated by superimposed migraine
Foggy, clouded or disoriented attacks, in which cases VM is not the primary culprit.
Drunk or seasick For example, migraine is more common in patients
Floating sensation, or walking on air or pillow with benign paroxysmal positional vertigo (BPPV)
Coming off a roller-coaster or merry-go-round than in age- and sex-matched controls (11,44,63–65),
Vestibular symptoms defined by ICVD and having migraine could be associated with an
Spontaneous vertigo as a false motion sensation of self or increased risk of developing BPPV (66). Vertigo can
surrounding
also be a feature of migraine with brainstem aura, but
Visually induced vertigo
Positional vertigo (after a change of head position)
other symptoms related to brainstem involvement (e.g.
Head motion-induced vertigo (during head motion) dysarthria) are typically not seen in VM patients.
Head motion-induced dizziness (sensation of disturbed spatial Thus, with such a wide range of clinical presentations,
orientation) the co-occurrence of migraine and vestibular symp-
toms is not specific to VM patients, and other dis-
orders with similar symptoms should always be
considered in the differential diagnosis (Figure 1). In
self-motion, or as external vertigo with primarily a this scheme, the distinction between VM and other
visual sensation of motion within surroundings. vestibular disorders such as Ménière’s disease can
Positional vertigo refers to a false sense of motion become unclear, especially since the auditory and ves-
after changing head position, and visually induced ver- tibular signs of labyrinthine pathology may be absent
tigo is characterized as a false sense of self-motion trig- in the early stages of Ménière’s disease (67,68).
gered by complex or large moving visual stimuli. Head Therefore, it is not surprising that both conditions
motion-induced vertigo refers to a distorted sense of are often reported to coincide (55,68–71). Migraine
motion during head movement. This is different from is more commonly reported in patients with
positional vertigo that occurs after a new head position Ménière’s disease than in otherwise healthy individ-
is adopted. Head motion-induced vertigo is also distin- uals, and about 13% of the patients meet the diagnos-
guished from motion sickness, in which the dominant tic criteria for both VM and Ménière’s disease (72).
symptom is nausea, and not vertigo. Various studies Such association has prompted theories about a link
have reported spontaneous vertigo in 20–85% of VM between these two conditions versus those that attri-
episodes, positional vertigo in 18–60%, and head bute the overlap to the secondary migraine symptoms
motion intolerance in 30–80% of the episodes provoked by vestibular symptoms in Ménière’s dis-
(5,6,9,36,39,44,52,53,56). These symptoms may vary ease. Whether there is a pathological link between
in individual patients, but spontaneous vertigo is the these two conditions remains unclear. Vestibular
most common reported vestibular symptom, followed symptoms in VM patients are also similar to those
by positional vertigo (12,57), head motion-induced ver- of another clinical entity that was recently defined as
tigo, and visually induced vertigo (5,58,59). The term persistent postural-perceptual dizziness (PPPD)
‘‘dizziness’’ is also included in the VM diagnostic cri- (73,74). Similar to VM, PPPD patients have symptoms
teria and is defined as a sense of disturbed spatial orien- consistent with perceptual dysfunction in spatial orien-
tation. However, it is not clear why the current VM tation and also experience fluctuating dizziness or
criteria only mention head motion-induced dizziness unsteadiness, provoked by postural changes or visual
with nausea, and other related terms such as spontan- motion. Both VM and PPPD patients may have
eous, positional, or visually induced dizziness are not comorbid psychiatric conditions such as anxiety and
included. depression (44,75–78). Also, migraine is one of the
The duration of vestibular symptoms in VM patients most common conditions associated with chronic
is quite variable (9,11,39,52). About 30% of patients visuospatial symptoms in PPPD patients (about
have symptoms lasting for minutes, 30% lasting for 15–20% of cases) (44,73). Since VM and PPPD are
hours, and 30% lasting for several days at a time. both defined only by clinical symptoms and have
The other 10% often report fluctuating daily symp- unknown pathogenesis, it is not clear whether the
toms. Nausea, vomiting, and susceptibility to motion overlap in their clinical presentations represents a
sickness are also frequent in VM patients; however, pathological link between these two conditions.
Huang et al. 111

Right head impulse Left head impulse


300 300
Covert Covert

Velocity [°/s]

Velocity [°/s]
saccades saccades
200 200

100 100

0 0

0 100 200 300 0 100 200 300


Time [ms] Time [ms]

Figure 1. Video head impulse testing (vHIT) shows bilateral vestibulopathy in a patient with clinical presentation consistent with
vestibular migraine (VM): 37 year old Navy man with 2-year history of fluctuating dizziness, described as feeling of being drunk lasting
for hours, and attacks with spinning sensation lasting for minutes without headaches. He reported frequent headaches, twice a month,
associated with dizziness, nausea, photophobia and phonophobia since age 25. Bedside neurological and vestibular examinations were
only remarkable for difficulty in tandem gait. The vHIT shows reduced vestibular gains in the horizontal canal planes at 0.7, calculated
as eye velocity (black traces) divided by head velocity (red traces: Right side and blue traces: Left side). There are also corrective
saccades in the eye traces. These ‘‘covert’’ saccades are embedded in the deficient vestibulo-ocular responses during head rotation and
are difficult to discern with the naked eye. Cases like this show that vestibular dysfunctions can be complicated by superimposed
migraine attacks; however, VM is not the primary culprit.

under Frenzel goggles). The nystagmus was low-velo-


Clinical finding and laboratory testing city and sustained during the vestibular attack, but it
Vestibular migraine is a diagnosis primarily based on dissipated during the symptom-free period.
clinical history, with currently no pathognomonic clin- While some studies have found a higher incidence of
ical sign or laboratory test that can verify its diagnosis. central vestibular dysfunction in VM patients (39,86),
Vestibular laboratory abnormalities are quite variable others have found no significant central vestibulo-
among VM patients, which may reflect inconsistent ocular findings (76,88), or have reported a higher inci-
findings regarding the existence of a peripheral vestibu- dence of peripheral vestibular dysfunction in VM
lar component. Nevertheless, vestibular testing is still patients (6,23,80,89). Such variability could be related
helpful to rule out other disorders considered in the to lack of standard diagnostic criteria among earlier
differential diagnosis (Figure 1). Neurological examin- studies or a sole reliance on clinical symptoms by the
ation is usually normal in VM patients during symp- current VM criteria, which does not prevent inclusion
tom-free periods. However, some mild, non-specific of patients with other vestibular disorders who may
vestibulo-ocular abnormalities have been reported in have superimposed migraine symptoms. In this context,
association with VM. Pursuit abnormality is reported findings such as asymmetry in caloric responses
in up to 48% of VM patients, spontaneous nystagmus (reported in up to 20% of VM patients), or vestibular
in about 10% of patients, and central positional loss with video head impulse testing (vHIT) cannot be
or gaze-evoked nystagmus in up to 28% of patients specific to VM and may represent a secondary migraine
(5–7,45,59,79–84). These non-specific ocular motor syndrome triggered by uncompensated vestibular dys-
abnormalities increased over time, from 16% to 41% function (Figure 1) (5,6,39,80,85,90–92).
of patients during a follow-up after 5.5 to 11 years (85). The cervical and ocular vestibular evoked myogenic
Apart from pursuit dysfunction, the most frequent potentials (cVEMP/oVEMP), the widely used labora-
ocular motor abnormality is central positional nystag- tory tests of otolith function, have also shown conflict-
mus. In a small study, about 70% of patients were ing results in VM patients. Some studies have reported
found to have pathological nystagmus during their reduced amplitude or delayed latencies of VEMP
VM attacks (86). Another study found nystagmus was responses (93–98), whereas other studies have found
provoked by horizontal headshaking in 35% of VM asymmetrical VEMP responses with normal latencies
patients, and spontaneous nystagmus in 19% of the and amplitudes (99,100). Three recent studies using
patients during vestibular attacks (87). Head shaking- ICHD criteria have reported abnormal oVEMP but
induced nystagmus is also reported in between the normal cVEMP responses in VM patients, which dif-
attacks (79,81). Vestibular migraine patients were also fered from the pattern of VEMP responses in normal
found to have positional nystagmus after removal of controls (101) or patients with Ménière’s disease. A sig-
visual fixation (e.g. vestibulo-ocular examination nificant vestibulo-ocular finding that could be related to
112 Cephalalgia 40(1)

Vestibular migraine Healthy control

Right
Right
80 80

Eye velocity °/sec


Eye velocity °/sec
0 0

Left
Left
–80 –80
0 10 20 30 0 10 20 30
Time Time
TC Right: 30 sec Gain right: 0.8 TC Right: 7 sec Gain right: 0.8
TC Left: 33 sec Gain left: 0.8 TC Left: 7 sec Gain left: 0.8

Figure 2. Vestibulo-ocular responses during step velocity rotations (60 /sec) in a VM patient versus a healthy control. Data points
show the slow phase velocity of vestibular nystagmus induced by rotation around the body axis to the right (positive values) and left
(negative values). The gains of vestibular responses (maximum eye velocity/60 /sec step velocity) are normal in both the VM patient
and the healthy control; however, the durations of nystagmus measured as time constant (TC) are longer in the VM patient compared
to the healthy control (30–33 sec versus 7 sec). See reference 106 for rotational chair testing.

VM pathophysiology is a longer duration of post-rota- pain or ‘‘central sensitization’’ that often presents as
tory nystagmus in VM patients compared with healthy allodynia (108–110). Some of these neurotransmitters
controls or migraine patients without dizziness (i.e. are also expressed in the vestibular system (e.g.
increased time constant of the vestibulo-ocular reflex) CGRP and serotonin) and might be involved in VM
(Figure 2) (81). VM patients were also found to have pathophysiology (108,111,112). The trigeminal nucleus
quicker modulation of the otolith-ocular response is connected to the contralateral thalamus, which in
during off-axis rotation (i.e. body centrifugation) (76). turn sends projections to the temporal, parietal, insular,
These findings suggest an innate hypersensitivity of the and cingulate cortical regions. The nociceptive brain-
vestibular system in these patients. Balance impairment stem centers such as the nucleus raphe magnus,
is also commonly reported in VM patients, including periaqueductal gray and hypothalamic areas are
abnormal Romberg and sensory organization test also connected with the TVS and vestibular nuclei
(SOT) (76,79,86,89,91,92). Studies using static posturo- (113,114,114). These reciprocal connections can modu-
graphy have found increased sway in VM patients com- late neural activity within both the TVS and the
pared with healthy controls (80,104). Hearing vestibular system (112,113,115–118). For example, tri-
impairment is reported in about 8% of VM patients, geminal stimulation produced nystagmus in migraine
which is often mild and non-progressive, as opposed to patients, suggesting increased vestibular excitability in
progressive low frequency hearing loss in Ménière’s these patients compared with healthy controls
patients (40,85,105). (116,119). Similarly, spontaneous nystagmus during
VM attacks or prolonged vestibular responses in
some VM patients (i.e. long time constant of vestibular
Pathophysiology nystagmus) could be linked to vestibular hyperexcit-
To date, VM pathogenesis remains obscure as the wide ability in these patients (76,81,120). It is, however, not
ranges of clinical and laboratory findings do not seem clear whether such hyperexcitability is at the level of the
consistent with a uniform pathology. Such variability peripheral vestibular system or the brainstem, or
comes from sole reliance on clinical symptoms and lack whether it could be related to the modulating effects
of objective measures for diagnosis, which have ham- of the cerebellum or cerebral hemispheres on the ves-
pered efforts toward understanding VM pathophysi- tibular system.
ology. Various mechanisms have been put forward to The phenomenon referred to as cortical spreading
explain VM symptoms and some are broadly inferred depression has been proposed as the neural correlate
from other migraine disorders (Figure 3). In this con- of migraine aura (107). This is a depolarization wave
text, altered neural activity within the trigeminovascu- that slowly spreads across the cerebral cortex, followed
lar system (TVS) is considered the primary mechanism by a prolonged suppression of cortical activity. The
for headache in migraine patients (107). The TVS theory of cortical spreading depression cannot explain
neuropeptides, such as substance P and calcitonin chronic or fluctuating symptoms in migraineurs.
gene-related peptide (CGRP), can cause vasodilation However, according to its proposed mechanism, symp-
and neurogenic inflammation, leading to throbbing toms such as hypersensitivity to light and sound could
Huang et al. 113

Thalamocortical Sensory dysmodulation


network Spatial disorientation

Nociceptive brainstem Trigemino-vascular


centers system Headache

Vestibular Vestibulo-ocular dysfunction


system Vestibular hypersensitivity

Figure 3. Possible mechanisms involved in the pathogenesis of vestibular migraine. Abnormal sensory modulation or integration
within the thalamo-cortical network could result in dizziness and spatial disorientation. Hyperactivity within the trigeminovascular
system (TVS) and nociceptive brainstem centers could result in headache. Altered activity in the vestibular system could lead to
transient vestibulo-ocular dysfunction or vestibular hypersensitivity associated with migraine features.

(a) (b) Dynamic roll tilt


VM patients 2
Control Normal
1.6 Migraine
Head upright Threshold
VM
1.2

0.8
*
0.4
Right head tilt
0
0.1 1
–4 0 4
Frequency (Hz)
L SVV R

Figure 4. (a) Spatial orientation measured as subjective visual vertical (SVV) errors in a group of VM patients versus healthy controls
(127). In the upright head position, there is no significant difference between VM patients and controls. During static lateral head tilt to
the right (20 ), SVV errors in VM patients are in the opposite direction of head tilt, consistent with overestimation of the head tilt
position for spatial orientation. (b) During low-frequency dynamic head tilt, VM patients show a lower motion detection threshold
compared with migraine patients without dizziness and healthy controls (modified with permission from Lewis et al. (125)).

be linked to a generally hyperexcitable brain state in reporting direction of rotation (126). During lateral
migraine patients (121). This has led to the concept of head tilts, VM patients had larger errors in spatial
sensory dysmodulation, in which deficient habituation orientation compared with healthy controls (Figure 4)
and potentiation of sensory responses are implicated in (127). When the head is tilted, the brain must integrate
migraine pathogenesis (122,123). Accordingly, expos- vestibular signals that encode head position with visual
ure to one sensory stimulus results in hypersensitivity inputs in order to maintain spatial orientation. The
that can extend to other sensory stimuli. VM patients, larger errors of spatial perception in VM patients
likewise, show higher sensitivity to motion in the roll were in the opposite direction of the head tilt, consist-
plane (e.g. lateral head tilt with respect to gravity) ent with overestimation of the head position in the pro-
(Figure 4) (120,124,125). Perception of tilt during cess of sensory integration for spatial orientation. In
whole body centrifugation is also altered in VM keeping with this result, VM patients reported dizziness
patients, suggesting that the integration of semicircular mostly in the same head direction that induced larger
canal and otolith inputs is affected in these patients. errors of spatial orientation. VM patients were also
During whole-body yaw rotations, VM patients were found to have high variability in spatial orientation
found to have altered perceptual thresholds while with the head in the upright position (128,129).
114 Cephalalgia 40(1)

Altogether, these findings suggest that visuospatial a role in VM patients warrants further research
symptoms in VM patients are related to altered sensory (48,145–150).
processing and integration that contribute to the per-
ception of spatial orientation. Regarding the effect of
Treatment
visual motion, VM patients had larger visual-induced
errors in spatial orientation during and after optoki- The uncertainties surrounding VM diagnosis over the
netic stimulation compared with healthy controls last few decades have also limited advances in treat-
(126,130,131). These errors also increased significantly ment. In general, VM patients can be managed with
in migraine patients with the duration of visual lifestyle modification, dietary adjustments, medications,
stimulation (132). In addition, VM patients had larger vestibular physical therapy, and activities that can
postural sway with visual motion compared with enhance perception of spatial orientation, such as
healthy controls (76,131,133). Regarding other sensory ping-pong or dancing. Given the considerable psycho-
modalities, there is evidence for sensory dysmodulation social impact of symptoms in VM patients, it is import-
affecting the auditory system in VM patients. In the ant to adjust treatment strategies based on each
presence of noise, otoacoustic emissions were less sup- patient’s needs. Those who are not significantly
pressed in VM patients compared with healthy con- impaired by their symptoms may benefit from reassur-
trols, suggesting a link to phonophobia in these ance to allay their fear and prevent unnecessary medical
patients (134). costs. This is especially important for patients with
Imaging studies suggest structural and functional prominent vestibular symptoms. Those who are signifi-
changes within the temporo-parietal regions in VM cantly impaired by their symptoms often benefit from
patients (117,135,136). The higher-order neural mech- combined interventions. In this process, identifying
anisms within these cortical regions are involved in triggers and implementing measures to circumvent
sensory integration for coherent spatial perception them can be used as the first line of treatment. As a
(137). In addition, VM patients were also found to key intervention, dietary adjustment and eliminating
have abnormal thalamic activities (117,138). These triggers such as red wine, aged cheeses, artificial sweet-
patients showed increased thalamic activations with eners, processed meats, chocolate, caffeine, MSG, and
cold water ear irrigation during fMRI studies, in com- alcohol are found to be effective in reducing symptoms
parison with migraine patients without aura or healthy (14,151,152).
controls (138). The magnitude of thalamic activation Like other migraine subtypes, medical treatment in
was positively correlated with the frequency of migraine VM patients aims to reduce frequency and severity of
attacks in VM patients. Also, increased ictal PET activ- symptoms. Most medications used for VM treatment
ities during vertigo attacks in VM patients were noted are aimed at prophylactic therapy. These medications
in the temporo-parieto-insular areas and bilateral tha- are mainly antihypertensives, antidepressants, and anti-
lami (117). These findings are in agreement with the epileptics with general applications in migraine preven-
studies that show the role of the thalamus as a major tion. Their use in VM treatment is supported by data
sensory relay within the vestibular pathways, and its mainly from small studies, and there is insufficient evi-
involvement in multisensory processing and integration dence from randomized controlled trials that can verify
including vestibular, visual, and somatosensory inputs the effect of these medications for VM prevention
(139,140). However, little is known about these sensory (Table 3) (153). Beta-adrenergic inhibitors such as pro-
processes and how these higher-level networks could be pranolol, calcium channel blockers such as verapamil,
affected in VM patients. and anti-epileptic medications such as topiramate
The familial occurrence of VM suggests a genetic and lamotrigine are sometimes effective in migraine
component in its pathogenesis. Although there are prophylaxis, but their role in VM treatment is not
some familial migraine syndromes such as hemiplegic clear (51,151,154–161). A prospective randomized
migraine or episodic ataxia, similar mutations (e.g. non-placebo-controlled study in VM patients suggested
CACNA1A or ATP1A2) are not found in VM patients flunarizine is effective in decreasing the severity and
(141,142). The CACNA1A gene encodes a subunit of frequency of vertigo attacks (159). A retrospective
the voltage-gated P/Q-type calcium channels, and its study found that cinnarizine decreased vertigo fre-
mutation can cause at least three neurological disorders quency in VM patients (160). Tricyclic antidepressants
linked to calcium channelopathy: Episodic ataxia type (TCAs), which have a broad mechanism of action, are
2, familial hemiplegic migraine type 1, and spinocere- shown to be effective in reducing episodic migraine
bellar ataxia type 6 (143). Vertigo and migraine symp- symptoms (154). Selective serotonin reuptake inhibitors
toms are also common in patients with episodic ataxia (SSRIs) or serotonin-norepinephrine reuptake inhibi-
type 2 (144). Despite the lack of evidence for mono- tors (SNRIs) have also been tried for VM prevention
genic inheritance, whether polygenic inheritance plays (154,161). These antidepressants may provide benefit in
Huang et al. 115

Table 3. Summary of studies on preventive treatments for vestibular migraine defined based on the ICHD-3 criteria.

Drug Daily dose Study design Duration Outcome

Propranolol 40–160 mg 33 patients, prospective, 4 months Severity: DHI 55.8  2.7 (Salviz et al. 2016)
randomized, controlled to 31.3  3.7* (158)
VSS 7.3  0.3 to 2.1  0.4*
Attacks per month: 12.6  1.8
to 1.9  0.7*
Venlafaxine 37.5–150 mg 31 patients, prospective, 4 months Severity: DHI 50.9  2.5 (Salviz et al. 2016)
randomized, controlled to 19.9  2.9* (158)
VSS 7.1  0.3 to 1.8  0.5*
Attacks per month: 12.2  1.8
to 2.6  1.1*
Venlafaxine 37.5 mg 25 patients, prospective 3 months Severity: DHI 41.7  16.9 (Liu et al. 2017)
to 31.3  14.1* (157)
VSS 5.9  1.7 to 3.8  1.2*
Flunarizine 10 mg 25 patients, prospective 3 months Severity: DHI 46.6  15.1 (Liu et al. 2017)
to 39.8  16.3* (157)
VSS 6.4  1.9 to 5.9  1.6*
Valproic acid 1000 mg 25 patients, prospective 3 months Severity: DHI 46.8  13.5 (Liu et al. 2017)
to 38.7  13.6* (157)
VSS 5.8  1.8 to 5.3  1.0
Cinnarizine 75 mg 24 patients, retrospective 3 months Attacks per month: 3.8  1.1 (Taghdiri et al. 2014)
open-label to 0.4  0.6* (160)
*p < 0.05.
DHI: dizziness handicap inventory (175); VSS: vertigo severity score (157,158).

cases with psychiatric comorbidities such as anxiety and effective in four patients with prolonged or frequent
depression. Recently, monoclonal anti-CGRP antibo- vertigo attacks in VM patients (167), similar to the
dies have been approved for migraine prevention. effect of steroids on patients with status migrainosus
Serum CGRP levels are elevated interictally in chronic (168).
migraine patients and to a lesser extent in those with Vestibular rehabilitation has been used to alleviate
episodic migraine (162). CGRP is also detected in symptoms and promote recovery in VM patients, as in
human cochlear and vestibular end organs, and thus other vestibular disorders (169). Although most studies
it may also play a role in vestibular physiology show that focused vestibular rehabilitation benefits
(163,164). However, it is unknown whether CGRP patients, no randomized control study has evaluated
has a role in VM pathogenesis and whether the the efficacy of vestibular physical therapy in VM
CGRP antagonists or monoclonal antibodies can be patients (170). One early study compared the effect
effective for VM treatment. of vestibular rehabilitation in 14 VM patients to
There are only a few randomized controlled studies 25 patients with migraine who had unrelated vestibular
on acute treatment for VM attacks. One small double- dysfunction, and showed both groups improved signifi-
blind, placebo-controlled study showed 38% of VM cantly on subjective and objective outcome measures
patients improved from severe or moderate vertigo to within four months (84). This finding was supported
mild or no vertigo in two hours after taking zolmitrip- by another study of 34 VM patients (14). A prospective
tan, compared to 22% with placebo (165). However, study in which 20 VM patients received a nine-week
due to the limited power of the study, the difference customized vestibular rehabilitation program also
did not reach statistical significance. Rizatriptan showed benefits, regardless of their medication regimen
reduced vestibular-induced motion sickness in migrain- (171). In another study, 28 VM patients based on
eurs compared to the placebo (166). The authors sug- ICHD-3 beta criteria and 79 patients with tension-
gested that rizatriptan may reduce motion sickness by type headache and dizziness were recruited (172).
influencing serotonergic vestibulo-autonomic path- Both groups were trained in vestibular therapy exer-
ways. Sumatriptan was also effective in improving cises during a five-day admission process and were
both headache and vertigo regardless of their temporal assessed 6 months later. The results showed that ves-
relation (155). Steroid injections were reported to be tibular rehabilitation contributed more prominently to
116 Cephalalgia 40(1)

clinical improvement in the VM group compared with diagnostic criteria. Neurological and ocular motor
the tension-type headache group. In practice, vestibular examination is usually normal in VM patients during
rehabilitation can be particularly beneficial to patients symptom-free periods. However, some mild, nonspeci-
if secondary complications such as deconditioning or fic vestibulo-ocular abnormalities such as positional
visual dependence have developed. VM patients often nystagmus have been reported, especially during VM
experience dizziness and disorientation with changes in attacks. So far, no pathognomonic clinical or labora-
head and body positions, which raises the possibility of tory finding exists for VM; however, vestibular testing
a ‘‘higher level’’ dysfunction in multisensory integration is still helpful to rule out other disorders considered in
for spatial orientation (i.e. visual, vestibular, and som- the differential diagnosis. Vestibular laboratory
atosensory inputs). Therefore, natural activities that abnormalities are quite variable among VM studies.
can enhance spatial perception and body coordination, Such variability could be related to the lack of standard
such as ping-pong and dancing, can be helpful to alle- diagnostic criteria used in earlier studies or the reliance
viate symptoms in these patients. Such an approach is on clinical symptoms alone in the current VM criteria,
supported by the effects of long-term training in discip- which does not prevent inclusion of patients with other
lines such as ballet dancing and yoga. Experts in these vestibular disorders who may have superimposed, sec-
fields are better at processing and weighting body per- ondary migraine symptoms. Like other types of
ceptual information and are less visually dependent in migraine, both abortive and preventive medications
their perception of spatial orientation (173,174). have been used in VM patients. However, the efficacy
of these medications has not been verified by large-
scale randomized placebo-controlled clinical trials.
Conclusions
Non-pharmacological treatment including life-style
Vestibular migraine is among the most common causes adjustment, trigger avoidance and vestibular rehabilita-
of recurrent vertigo in the general population, with lar- tion are also shown to be beneficial in VM patients. The
gely unknown pathophysiology. Patients with VM pathophysiology of VM remains largely unknown;
experience disabling spatial misperceptions including however, recent findings have provided some prelimin-
unusual sensitivity to head motion or visual stimuli, ary insights. These include reports of abnormal motion-
or sudden feelings of imbalance or tilt. These symptoms detection thresholds during roll tilt or yaw rotation, or
often become chronic and lead to severe impairment of larger errors of spatial orientation during head tilt,
daily activities in VM sufferers. Unlike typical migraine which suggest high-level dysfunctions related to ves-
aura, vestibular symptoms in VM patients are not tem- tibular processing in these patients. So far there is no
porary symptoms that occur with headaches, and their information about the plausible role of central neural
duration can last from hours to several days. Currently, processes in VM spatial disorientation and their link to
the VM diagnosis is entirely based on clinical his- visuospatial symptoms in these patients. Therefore,
tory, and according to the recent consensus of the gaining insight into these processes and how they
International Headache and the Bárány Societies, the result in dizziness, vertigo, and spatial disorientation
presence of episodic vestibular symptoms associated in VM patients is an important step towards devising
with at least one migraine feature can fulfill the VM effective treatment strategies.

Clinical implications
. VM is among the most common causes of recurrent vertigo. In addition to vertigo, patients may experience
disabling spatial misperceptions.
. The diagnosis of VM is entirely based on clinical history, and no laboratory test is pathognomonic.
. The pathophysiology of VM remains obscure, but recent studies suggest sensory dysfunction is related to
processing vestibular inputs.
. The treatment of VM requires lifestyle adjustment, trigger avoidance and vestibular rehabilitation as well as
abortive and preventive medications, although solid evidence is still lacking.

Declaration of conflicting interests Funding


The authors declared no potential conflicts of interest with The authors disclosed receipt of the following financial sup-
respect to the research, authorship, and/or publication of this port for the research, authorship, and/or publication of this
article. article: This study was supported in part by the following
Huang et al. 117

grants: Ministry of Science and Technology of Taiwan 15. Vuković V, Plavec D, Galinović I, et al. Prevalence of
[MOST 107-2321-B-010-001], Brain Research Center, vertigo, dizziness, and migrainous vertigo in patients
National Yang-Ming University from The Featured Areas with migraine. Headache 2007; 47: 1427–1435.
Research Center Program within the framework of the 16. Akdal G, Özge A and Ergör G. The prevalence of ves-
Higher Education Sprout Project by the Ministry of tibular symptoms in migraine or tension-type headache.
Education (MOE) in Taiwan, as well as the United States J Vestib Res: Equilib Orient 2013; 23: 101–106.
National Institute of Deafness and Other Communication 17. Eggers SDZ, Staab JP, Neff BA, et al. Investigation of
Disorders (NIDCD); K23DC013552. the coherence of definite and probable vestibular
migraine as distinct clinical entities. Otol Neurotol 2011;
ORCID iDs 32: 1144–1151.
18. Aragones JM, Fortes-Rego J, Fuste J, et al. Migraine:
Tzu-Chou Huang https://orcid.org/0000-0003-4674-7882
An alternative in the diagnosis of unclassified vertigo.
Shuu-Jiun Wang https://orcid.org/0000-0001-5179-5358
Headache 1993; 33: 125–128.
19. Cha Y-H, Lee H, Santell LS, et al. Association of benign
References recurrent vertigo and migraine in 208 patients.
1. Stovner LJ, Nichols E, Steiner TJ, et al. Global, regional, Cephalalgia 2009; 29: 550–555.
and national burden of migraine and tension-type head- 20. Lee H, Il Sohn S, Kyo Jung D, et al. Migraine and iso-
ache, 1990–2016: A systematic analysis for the Global lated recurrent vertigo of unknown cause. Neurol Res
Burden of Disease Study 2016. Lancet Neurol 2018; 17: 2002; 24: 663–665.
954–976. 21. Power L, Shute W, McOwan B, et al. Clinical character-
2. Bösner S, Schwarm S, Grevenrath P, et al. Prevalence, istics and treatment choice in vestibular migraine. J Clin
aetiologies and prognosis of the symptom dizziness in Neurosci 2018; 52: 50–53.
primary care – a systematic review. BMC Fam Pract 22. Rassekh CH and Harker LA. The prevalence of
2018; 19: 33. DOI: 10.1186/s12875-017-0695-0. migraine in Menière’s disease. Laryngoscope 1992; 102:
3. Newman-Toker DE, Hsieh Y-H, Camargo CA, et al. 135–138.
Spectrum of dizziness visits to US emergency 23. Savundra P, Carroll J, Davies R, et al. Migraine-
departments: Cross-sectional analysis from a nationally associated vertigo. Cephalalgia 2002; 17: 505–510.
representative sample. Mayo Clin Proc 2008; 83: 765–775. 24. Neuhauser H and Lempert T. Vertigo and dizziness
4. Calhoun AH, Ford S, Pruitt AP, et al. The point related to migraine: A diagnostic challenge. Cephalalgia
prevalence of dizziness or vertigo in migraine – and 2004; 24: 83–91.
factors that influence presentation. Headache 2011; 51: 25. Headache Classification Committee of the International
1388–1392. Headache Society (IHS). The International Classification
5. Cass SP, Ankerstjerne JKP, Yetiser S, et al. Migraine- of Headache Disorders, 3rd edition. Cephalalgia 2018; 38:
related vestibulopathy. Ann Otol Rhinol Laryngol 1997; 1–211.
106: 182–189. 26. Lempert T, Olesen J, Furman J, et al. Vestibular
6. Cutrer FM and Baloh RW. Migraine-associated dizzi- migraine: Diagnostic criteria. J Vestib Res 2012; 22:
ness. Headache 1992; 32: 300–304. 167–172.
7. Dieterich M and Brandt T. Episodic vertigo related to 27. Radtke A, Neuhauser H, von Brevern M, et al. Vestibular
migraine (90 cases): Vestibular migraine? J Neurol 1999; migraine – validity of clinical diagnostic criteria.
246: 883–892. Cephalalgia 2011; 31: 906–913.
8. Johnson GD. Medical management of migraine-related 28. Bisdorff A, Von Brevern M, Lempert T, et al. Classifica-
dizziness and vertigo. Laryngoscope 1998; 108: 1–28. tion of vestibular symptoms: Towards an international
9. Kayan A and Hood JD. Neuro-otological manifestations classification of vestibular disorders. J Vestib Res 2009;
of migraine. Brain 1984; 107: 1123–1142. 19: 1–13.
10. Kuritzky A, Ziegler DK and Hassanein R. Vertigo, 29. Newman-Toker DE, Cannon LM, Stofferahn ME, et al.
motion sickness and migraine. Headache 1981; 21: Imprecision in patient reports of dizziness symptom qual-
227–231. ity: A cross-sectional study conducted in an acute care
11. Neuhauser H, Leopold M, Von Brevern M, et al. The setting. Mayo Clin Proc 2007; 82: 1329–1340.
interrelations of migraine, vertigo, and migrainous ver- 30. Radtke A, von Brevern M, Neuhauser H, et al. Vestibular
tigo. Neurology 2001; 56: 436–441. migraine: Long-term follow-up of clinical symptoms and
12. Neuhauser HK, Radtke A, Von Brevern M, et al. Migrai- vestibulo-cochlear findings. Neurology 2012; 79:
nous vertigo prevalence and impact on quality of life. 1607–1614.
Neurology 2006; 67: 1028–1033. 31. To-Alemanji J, Ryan C and Schubert MC. Experiences
13. Oh AK, Lee H, Jen JC, et al. Familial benign recurrent engaging healthcare when dizzy. Otol Neurotol 2016; 37:
vertigo. Am J Medical Genetics 2001; 100: 287–291. 1122–1127.
14. Reploeg MD and Goebel JA. Migraine-associated dizzi- 32. Batu ED, Anlar B, Topçu M, et al. Vertigo in childhood:
ness: Patient characteristics and management options. A retrospective series of 100 children. Eur J Paediatr
Otol Neurotol 2002; 23: 364–371. Neurol 2015; 19: 226–232.
118 Cephalalgia 40(1)

33. Brandt T and Strupp M. Migraine and vertigo: Classifi- 52. Zhang Y, Kong Q, Chen J, et al. International Classifica-
cation, clinical features, and special treatment consider- tion of Headache Disorders 3rd edition beta-based field
ations. Headache Curr 2006; 3: 12–19. testing of vestibular migraine in China: Demographic,
34. Jahn K, Langhagen T and Heinen F. Vertigo and dizzi- clinical characteristics, audiometric findings and diagno-
ness in children. Curr Opin Neurol 2015; 28: 78–82. sis statues. Cephalalgia 2016; 36: 240–248.
35. Abu-Arafeh I and Russell G. Paroxysmal vertigo as a 53. Cohen JM, Bigal ME and Newman LC. Migraine and
migraine equivalent in children: A population-based vestibular symptoms – identifying clinical features that
study. Cephalalgia 1995; 15: 22–25, discussion 4. predict ‘‘vestibular migraine.’’ Headache 2011; 51:
36. Teggi R, Colombo B, Albera R, et al. Clinical features, 1393–1397.
familial history, and migraine precursors in patients with 54. Kırkım G, Mutlu B, Olgun Y, et al. Comparison of
definite vestibular migraine: The VM-Phenotypes pro- audiological findings in patients with vestibular migraine
jects. Headache 2018; 58: 534–544. and migraine. Turk Arch Otorhinolaryngol 2017; 55:
37. Formeister EJ, Rizk HG, Kohn MA, et al. The epidemi- 158–161.
ology of vestibular migraine: A population-based survey 55. Neff BA, Staab JP, Eggers SD, et al. Auditory and ves-
study. Otol Neurotol 2018; 39: 1037–1044. tibular symptoms and chronic subjective dizziness in
38. Hsu L-C, Wang S-J and Fuh J-L. Prevalence and impact patients with Ménière’s disease, vestibular migraine, and
of migrainous vertigo in mid-life women: A community- Ménière’s Disease with concomitant vestibular migraine.
based study. Cephalalgia 2011; 31: 77–83. Otol Neurotol 2012; 33: 1235–1244.
39. Dieterich M and Brandt T. Episodic vertigo related to 56. Dieterich M, Staab JP and Brandt T. Functional (psycho-
migraine (90 cases): Vestibular migraine? J Neurol 1999; genic) dizziness. Handb Clin Neurol 2016; 139: 447–468.
246: 883–892. 57. Dieterich M, Obermann M and Celebisoy N. Vestibular
40. Lempert T and Neuhauser H. Epidemiology of vertigo, migraine: The most frequent entity of episodic vertigo.
migraine and vestibular migraine. J Neurol 2009; 256: J Neurol 2016; 263: 82–89.
333–338. 58. Waterston J. Chronic migrainous vertigo. J Clin Neurosci
41. Ombergen AV, Rompaey VV, de Heyning PV, et al. Ves- 2004; 11: 384–388.
tibular migraine in an otolaryngology clinic: Prevalence, 59. Radtke A, von Brevern M, Neuhauser H, et al. Vestibular
associated symptoms, and prophylactic medication effect- migraine: Long-term follow-up of clinical symptoms and
iveness. Otol Neurotol 2015; 36: 133–138. vestibulo-cochlear findings. Neurology 2012; 79:
42. Geser R and Straumann D. Referral and final diag- 1607–1614.
noses of patients assessed in an academic vertigo center. 60. Drummond PD. Triggers of motion sickness in migraine
Front Neurol 2012; 3: 169. DOI: 10.3389/ sufferers. Headache 2005; 45: 653–656.
fneur.2012.00169. 61. Marcus DA, Furman JM and Balaban CD. Motion sick-
43. Cho S-J, Kim B-K, Kim B-S, et al. Vestibular migraine in ness in migraine sufferers. Expert Opin Pharmacother
multicenter neurology clinics according to the appendix cri- 2005; 6: 2691–2697.
teria in the third beta edition of the International Classifica- 62. Vitkovic J, Paine M and Rance G. Neuro-otological find-
tion of Headache Disorders. Cephalalgia 2016; 36: 454–462. ings in patients with migraine- and nonmigraine-related
44. Beh SC, Masrour S, Smith SV, et al. The spectrum of dizziness. Audiol Neurootol 2008; 13: 113–122.
vestibular migraine: Clinical features, triggers, and exam- 63. Bath AP, Walsh RM, Ranalli P, et al. Experience from a
ination findings. Headache 2019; 59: 727–740. multidisciplinary ‘‘dizzy’’ clinic. Am J Otol 2000; 21: 92–97.
45. Casani AP, Sellari-Franceschini S, Napolitano A, et al. 64. Ishiyama A, Jacobson KM and Baloh RW. Migraine and
Otoneurologic dysfunctions in migraine patients with or benign positional vertigo. Ann Otol Rhinol Laryngol 2000;
without vertigo. Otol Neurotol 2009; 30: 961–967. 109: 377–380.
46. Thakar A, Anjaneyulu C and Deka RC. Vertigo syn- 65. Lempert T, Leopold M, von Brevern M, et al. Migraine
dromes and mechanisms in migraine. J Laryngol Otol and benign positional vertigo. Ann Otol Rhinol Laryngol
2001; 115: 782–787. 2000; 109: 1176.
47. Park JH and Viirre E. Vestibular migraine may be an 66. Chu C-H, Liu C-J, Lin L-Y, et al. Migraine is associated
important cause of dizziness/vertigo in perimenopausal with an increased risk for benign paroxysmal pos-
period. Med Hypoth 2010; 75: 409–414. itional vertigo: A nationwide population-based study.
48. Lee H, Jen JC, Cha Y-H, et al. Phenotypic and genetic J Headache Pain 2015; 16: 62. DOI: 10.1186/s10194-
analysis of a large family with migraine-associated ver- 015-0547-z.
tigo. Headache 2008; 48: 1460–1467. 67. Gürkov R, Jerin C, Flatz W, et al. Clinical manifestations
49. Cha Y-H and Cui Y. Rocking dizziness and headache: A of hydropic ear disease (Menière’s). Eur Arch
two-way street. Cephalalgia 2013; 33: 1160–1169. Otorhinolaryngol 2019; 276: 27–40.
50. Andress-Rothrock D, King W and Rothrock J. An ana- 68. Radtke A, Lempert T, Gresty MA, et al. Migraine and
lysis of migraine triggers in a clinic-based population: Ménière’s disease: Is there a link? Neurology 2002; 59:
September 2010. Headache 2010; 50: 1366–1370. 1700–1704.
51. Baier B, Winkenwerder E and Dieterich M. ‘‘Vestibular 69. Brantberg K and Baloh RW. Similarity of vertigo attacks
migraine’’: Effects of prophylactic therapy with various due to Meniere’s disease and benign recurrent vertigo,
drugs: A retrospective study. J Neurol 2009; 256: both with and without migraine. Acta Otolaryngol 2011;
436–442. 131: 722–727.
Huang et al. 119

70. Ghavami Y, Mahboubi H, Yau AY, et al. Migraine fea- 88. Bir LS, Ardiç FN, Kara CO, et al. Migraine patients
tures in patients with Meniere’s disease. Laryngoscope with or without vertigo: Comparison of clinical and elec-
2016; 126: 163–168. tronystagmographic findings. J Otolaryngol 2003; 32:
71. Lopez-Escamez JA, Dlugaiczyk J, Jacobs J, et al. Accom- 234–238.
panying symptoms overlap during attacks in Menière’s 89. Teggi R, Colombo B, Bernasconi L, et al. Migrainous
disease and vestibular migraine. Front Neurol 2014; 5: 265. vertigo: Results of caloric testing and stabilometric find-
72. Cha Y-H, Kane MJ and Baloh RW. Familial clustering ings. Headache 2009; 49: 435–444.
of migraine, episodic vertigo, and Ménière’s disease. Otol 90. Blödow A, Heinze M, Bloching MB, et al. Caloric
Neurot 2008; 29: 93–96. stimulation and video-head impulse testing in Ménière’s
73. Staab JP, Eckhardt-Henn A, Horii A, et al. Diagnostic disease and vestibular migraine. Acta Otolaryngol 2014;
criteria for persistent postural-perceptual dizziness 134: 1239–1244.
(PPPD): Consensus document of the committee for the 91. Harno H, Hirvonen T, Kaunisto MA, et al. Subclinical
Classification of Vestibular Disorders of the Bárány vestibulocerebellar dysfunction in migraine with and
Society. J Vestib Res 2017; 27: 191–208. without aura. Neurology 2003; 61: 1748–1752.
74. Staab JP and Ruckenstein MJ. Expanding the differential 92. Kang WS, Lee SH, Yang CJ, et al. Vestibular function
diagnosis of chronic dizziness. Arch Otolaryngol Head tests for vestibular migraine: Clinical implication of
Neck Surg 2007; 133: 170–176. video head impulse and caloric tests. Front Neurol
75. Best C, Tschan R, Eckhardt-Henn A, et al. Who is at risk 2016; 7: 166.
for ongoing dizziness and psychological strain after a ves- 93. Baier B, Stieber N and Dieterich M. Vestibular-evoked
tibular disorder? Neuroscience 2009; 164: 1579–1587. myogenic potentials in vestibular migraine. J Neurol
76. Furman JM, Sparto PJ, Soso M, et al. Vestibular func- 2009; 256: 1447–1454.
tion in migraine-related dizziness: A pilot study. J Vestib 94. Baier B and Dieterich M. Vestibular-evoked myogenic
Res 2005; 15: 327–332. potentials in ‘‘vestibular migraine’’ and Menière’s dis-
77. Honaker JA, Gilbert JM, Shepard NT, et al. Adverse ease: A sign of an electrophysiological link? Ann NY
effects of health anxiety on management of a patient Acad Sci 2009; 1164: 324–327.
with benign paroxysmal positional vertigo, vestibular 95. Boldingh MI, Ljostad U, Mygland A, et al. Vestibular
migraine and chronic subjective dizziness. Am J sensitivity in vestibular migraine: VEMPs and motion
Otolaryngol 2013; 34: 592–595. sickness susceptibility. Cephalalgia 2011; 31:
78. Popkirov S, Staab JP and Stone J. Persistent postural- 1211–1219.
perceptual dizziness (PPPD): A common, characteristic 96. Murofushi T, Ozeki H, Inoue A, et al. Does migraine-
and treatable cause of chronic dizziness. Pract Neurol associated vertigo share a common pathophysiology
2018; 18: 5–13. with Meniere’s disease? Study with vestibular-evoked
79. Boldingh MI, Ljøstad U, Mygland Å, et al. Comparison myogenic potential. Cephalalgia 2009; 29: 1259–1266.
of interictal vestibular function in vestibular migraine vs 97. Roceanu A, Allena M, De Pasqua V, et al. Abnormal-
migraine without vertigo. Headache 2013; 53: 1123–1133. ities of the vestibulo-collic reflex are similar in migrain-
80. Çelebisoy N, Gökçay F, Şirin H, et al. Migrainous ver- eurs with and without vertigo. Cephalalgia 2008; 28:
tigo: Clinical, oculographic and posturographic findings. 988–990.
Cephalalgia 2008; 28: 72–77. 98. Zuniga MG, Janky KL, Schubert MC, et al. Can ves-
81. Jeong S-H, Oh S-Y, Kim H-J, et al. Vestibular dysfunc- tibular-evoked myogenic potentials help differentiate
tion in migraine: Effects of associated vertigo and motion Meniere disease from vestibular migraine? Otolaryngol
sickness. J Neurol 2010; 257: 905–912. Head Neck Surg 2012; 146: 788–796.
82. von Brevern M. Vestibular migraine: Vestibular testing and 99. Kandemir A, Çelebisoy N and Köse T. Cervical vestibu-
pathophysiology. In: Colombo B and Teggi R (eds) Ves- lar evoked myogenic potentials in primary headache dis-
tibular migraine and related syndromes. Cham, Switzerland: orders. Clin Neurophysiol 2013; 124: 779–784.
Springer International Publishing, 2014, pp.83–90. 100. Taylor RL, Zagami AS, Gibson WP, et al. Vestibular
83. von Brevern M, Radtke A, Clarke AH, et al. Migrainous evoked myogenic potentials to sound and vibration:
vertigo presenting as episodic positional vertigo. Characteristics in vestibular migraine that enable separ-
Neurology 2004; 62: 469–472. ation from Menière’s disease. Cephalalgia 2012; 32:
84. Whitney SL, Wrisley DM, Brown KE, et al. Physical 213–225.
therapy for migraine-related vestibulopathy and vestibu- 101. Zaleski A, Bogle J, Starling A, et al. Vestibular evoked
lar dysfunction with history of migraine. Laryngoscope myogenic potentials in patients with vestibular migraine.
2000; 110: 1528–1534. Otol Neurotol 2015; 36: 295–302.
85. Radtke A, von Brevern M, Neuhauser H, et al. Vestibular 102. Inoue A, Egami N, Fujimoto C, et al. Vestibular evoked
migraine: Long-term follow-up of clinical symptoms and myogenic potentials in vestibular migraine: Do they help
vestibulo-cochlear findings. Neurology 2012; 79: 1607–1614. differentiating from Menière’s disease? Ann Otol Rhinol
86. von Brevern M. Acute migrainous vertigo: Clinical and Laryngol 2016; 125: 931–937.
oculographic findings. Brain 2004; 128: 365–374. 103. Makowiec KF, Piker EG, Jacobson GP, et al. Ocular
87. Polensek SH and Tusa RJ. Nystagmus during attacks of and cervical vestibular evoked myogenic potentials in
vestibular migraine: An aid in diagnosis. Audiol Neurotol patients with vestibular migraine. Otol Neurotol 2018;
2010; 15: 241–246. 39: e561–e567.
120 Cephalalgia 40(1)

104. Gorski LP, da Silva AM, Cusin FS, et al. Body balance 123. Schwedt TJ. Multisensory integration in migraine. Curr
at static posturography in vestibular migraine. Braz J Opin Neurol 2013; 26: 248–253.
Otorhinolaryngol 2019; 85: 183–192. 124. Lewis RF, Priesol AJ, Nicoucar K, et al. Dynamic tilt
105. Battista RA. Audiometric findings of patients with thresholds are reduced in vestibular migraine. J Vestib
migraine-associated dizziness. Otol Neurotol 2004; 25: Res 2011; 21: 323–330.
987–992. 125. Lewis RF, Priesol AJ, Nicoucar K, et al. Abnormal
106. Rambold HA. Clinical value of rotational-chair testing motion perception in vestibular migraine.
in vestibular disease. Clin of Otorhinolaryngol 2017; 1: Laryngoscope 2011; 121: 1124–1125.
013. 126. Bednarczuk NF, Bonsu A, Ortega MC, et al. Abnormal
107. Pietrobon D and Moskowitz MA. Pathophysiology of visuo-vestibular interactions in vestibular migraine:
migraine. Annu Rev Physiol 2013; 75: 365–391. A cross sectional study. Brain 2019; 142: 606–616.
108. Durham PL. Calcitonin gene-related peptide (CGRP) 127. Winnick A, Sadeghpour S, Otero-Millan J, et al. Errors
and migraine. Headache 2006; 46: S3–S8. of upright perception in patients with vestibular
109. Furman JM, Marcus DA and Balaban CD. Migrainous migraine. Front Neurol 2018; 9: 892.
vertigo: Development of a pathogenetic model and struc- 128. Asai M, Aoki M, Hayashi H, et al. Subclinical deviation
tured diagnostic interview. Curr Opin Neurol 2003; 16: of the subjective visual vertical in patients affected by a
5–13. primary headache. Acta Otolaryngol 2009; 129: 30–35.
110. Schytz HW, Olesen J and Ashina M. The PACAP recep- 129. Kandemir A, Çelebisoy N and Köse T. Perception of
tor: A novel target for migraine treatment. verticality in patients with primary headache disorders.
Neurotherapeutics 2010; 7: 191–196. J Int Adv Otol 2014; 10: 138–143.
111. Ahn S-K and Balaban CD. Distribution of 5-HT1B and 130. Ashish G, Augustine AM, Tyagi AK, et al. Subjective
5-HT1D receptors in the inner ear. Brain Res 2010; visual vertical and horizontal in vestibular migraine.
1346: 92–101. J Int Adv Otol 2017; 13: 254–258.
112. Koo J-W and Balaban C. Serotonin-induced plasma 131. Guerraz M, Yardley L, Bertholon P, et al. Visual ver-
extravasation in the murine inner ear: Possible mechan- tigo: Symptom assessment, spatial orientation and pos-
ism of migraine-associated inner ear dysfunction. tural control. Brain 2001; 124: 1646–1656.
Cephalalgia 2006; 26: 1310–1319. 132. Miller MA and Crane BT. Roll vection in migraine and
113. Akerman S, Holland PR and Goadsby PJ. Diencephalic controls using inertial nulling and certainty estimate
and brainstem mechanisms in migraine. Nat Rev techniques. PLoS One 2017; 12(2): e0171332. DOI:
Neurosci 2011; 12: 570–584. 10.1371/journal.pone.0171332.
114. Halberstadt AL and Balaban CD. Organization of pro- 133. Lim Y-H, Kim J-S, Lee H-W, et al. Postural instability
jections from the raphe nuclei to the vestibular nuclei in induced by visual motion stimuli in patients with ves-
rats. Neuroscience 2003; 120: 573–594. tibular migraine. Front Neurol 2018; 9: 433.
115. Furman JM, Marcus DA and Balaban CD. Vestibular 134. Murdin L, Premachandra P and Davies R. Sensory dys-
migraine: Clinical aspects and pathophysiology. Lancet modulation in vestibular migraine: An otoacoustic emis-
Neurol 2013; 12: 706–715. sion suppression study. Laryngoscope 2010; 120:
116. Marano E, Marcelli V, Stasio ED, et al. Trigeminal 1632–1636.
stimulation elicits a peripheral vestibular imbalance in 135. Demarquay G, Ducros A, Montavont A, et al. Migraine
migraine patients. Headache 2005; 45: 325–331. with brainstem aura: Why not a cortical origin?
117. Shin JH, Kim YK, Kim H-J, et al. Altered brain Cephalalgia 2018; 38: 1687–1695.
metabolism in vestibular migraine: Comparison 136. Obermann M, Wurthmann S, Steinberg BS, et al. Cen-
of interictal and ictal findings. Cephalalgia 2014; 34: tral vestibular system modulation in vestibular migraine.
58–67. Cephalalgia 2014; 34: 1053–1061.
118. Weiller C, May A, Limmroth V, et al. Brain stem acti- 137. Kheradmand A and Winnick A. Perception of upright:
vation in spontaneous human migraine attacks. Nat Multisensory convergence and the role of temporo-par-
Med 1995; 1: 658–660. ietal cortex. Front Neurol 2017; 8: 552.
119. Vass Z, Steyger PS, Hordichok AJ, et al. Capsaicin stimu- 138. Russo A, Marcelli V, Esposito F, et al. Abnormal thal-
lation of the cochlea and electric stimulation of the tri- amic function in patients with vestibular migraine.
geminal ganglion mediate vascular permeability in Neurology 2014; 82: 2120–2126.
cochlear and vertebro-basilar arteries: A potential cause 139. Lopez C and Blanke O. The thalamocortical vestibular
of inner ear dysfunction in headache. Neuroscience 2001; system in animals and humans. Brain Res Rev 2011; 67:
103: 189–201. 119–146.
120. King S, Wang J, Priesol AJ, et al. Central integration of 140. Wijesinghe R, Protti DA and Camp AJ. Vestibular
canal and otolith signals is abnormal in vestibular interactions in the thalamus. Front Neural Circuits
migraine. Front Neurol 2014; 5: 233. 2015; 9: 79.
121. Aurora S and Wilkinson F. The brain is hyperexcitable 141. Kim J-S, Yue Q, Jen JC, et al. Familial migraine with
in migraine. Cephalalgia 2007; 27: 1442–1453. vertigo: No mutations found in CACNA1A. Am J Med
122. Goadsby PJ, Holland PR, Martins-Oliveira M, et al. Genetics 1998; 79: 148–151.
Pathophysiology of migraine: A disorder of sensory pro- 142. von Brevern M, Ta N, Shankar A, et al. Migrainous
cessing. Physiol Rev 2017; 97: 553–622. vertigo: Mutation analysis of the candidate genes
Huang et al. 121

CACNA1A, ATP1A2, SCN1A, and CACNB4. 159. Lepcha A, Amalanathan S, Augustine AM, et al. Flu-
Headache 2006; 46: 1136–1141. narizine in the prophylaxis of migrainous vertigo: A
143. Requena T, Espinosa-Sanchez JM and Lopez-Escamez randomized controlled trial. Euro Arch Oto Rhino
JA. Genetics of dizziness: Cerebellar and vestibular dis- Laryngol 2014; 271: 2931–2936.
orders. Curr Opin Neurol 2014; 27: 98–104. 160. Taghdiri F, Togha M, Razeghi Jahromi S, et al. Cinnar-
144. Jen JC, Graves TD, Hess EJ, et al. Primary episodic izine for the prophylaxis of migraine associated vertigo:
ataxias: Diagnosis, pathogenesis and treatment. Brain A retrospective study. SpringerPlus 2014; 3: 231.
2007; 130: 2484–2493. 161. Bisdorff AR. Management of vestibular migraine. Ther
145. Bahmad F, DePalma SR, Merchant SN, et al. Locus for Adv Neurol Disord 2011; 4: 183–191.
familial migrainous vertigo disease maps to chromo- 162. Cernuda-Morollón E, Larrosa D, Ramón C, et al. Inter-
some 5q35. Ann Otol Rhinol Laryngol 2009; 118: ictal increase of CGRP levels in peripheral blood as a
670–676. biomarker for chronic migraine. Neurology 2013; 81:
146. Baloh RW. Migraine and vertigo: Epidemiology, gen- 1191–1196.
etics, and mechanism(s). Headache Curr 2006; 3: 1–7. 163. Ramón C, Cernuda-Morollón E and Pascual J. Calcito-
147. Lea RA, Shepherd AG, Curtain RP, et al. A typical nin gene-related peptide in peripheral blood as a bio-
migraine susceptibility region localizes to chromosome marker for migraine. Curr Opin Neurol 2017; 30:
1q31. Neurogenetics 2002; 4: 17–22. 281–286.
148. Lee H, Jen JC, Wang H, et al. A genome-wide linkage 164. Schrott-Fischer A, Kammen-Jolly K, Scholtz A, et al.
scan of familial benign recurrent vertigo: Linkage to Efferent neurotransmitters in the human cochlea and
22q12 with evidence of heterogeneity. Hum Mol Genet vestibule. Acta Otolaryngol 2007; 127: 13–19.
2006; 15: 251–258. 165. Neuhauser H, Radtke A, von Brevern M, et al. Zolmi-
149. Nyholt DR, Morley KI, Ferreira MAR, et al. Genome- triptan for treatment of migrainous vertigo: A pilot ran-
wide significant linkage to migrainous headache on domized placebo-controlled trial. Neurology 2003; 60:
chromosome 5q21. Am J Hum Genet 2005; 77: 500–512. 882–883.
150. Wessman M, Kallela M, Kaunisto MA, et al. A suscep- 166. Furman JM, Marcus DA and Balaban CD. Rizatriptan
tibility locus for migraine with aura, on chromosome reduces vestibular-induced motion sickness in migrain-
4q24. Am J Hum Genet 2002; 70: 652–662. eurs. J Headache Pain 2011; 12: 81–88.
151. Maione A. Migraine-related vertigo: Diagnostic criteria 167. Prakash S and Shah ND. Migrainous vertigo responsive
and prophylactic treatment. Laryngoscope 2006; 116: to intravenous methylprednisolone: Case reports.
1782–1786. Headache 2009; 49: 1235–1239.
152. Mikulec AA, Faraji F and Kinsella LJ. Evaluation of 168. Rozen TD. Migraine headache: Immunosuppressant
the efficacy of caffeine cessation, nortriptyline, and therapy. Curr Treat Options Neurol 2002; 4: 395–401.
topiramate therapy in vestibular migraine and complex 169. Whitney SL, Alghadir AH and Anwer S. Recent evi-
dizziness of unknown etiology. Am J Otolaryngol 2012; dence about the effectiveness of vestibular rehabilita-
33: 121–127. tion. Curr Treat Options Neurol 2016; 18: 13.
153. Fernández MM, Birdi JS, Irving GJ, et al. Pharmacolo- 170. Alghadir AH and Anwer S. Effects of vestibular
gical agents for the prevention of vestibular migraine. rehabilitation in the management of a vestibular
Cochrane Database Syst Rev 2015; (6): CD010600. migraine: A review. Front Neurol 2018; 9: 440.
DOI: 10.1002/14651858.CD010600.pub2. 171. Vitkovic J, Winoto A, Rance G, et al. Vestibular
154. Jackson JL, Cogbill E, Santana-Davila R, et al. A com- rehabilitation outcomes in patients with and without
parative effectiveness meta-analysis of drugs for the vestibular migraine. J Neurol 2013; 260: 3039–3048.
prophylaxis of migraine headache. PLoS One 2015; 10: 172. Sugaya N, Arai M and Goto F. Is the headache in
e0130733. patients with vestibular migraine attenuated by vestibu-
155. Bikhazi P, Jackson C and Ruckenstein MJ. Efficacy of lar rehabilitation? Front Neurol 2017; 8: 124.
antimigrainous therapy in the treatment of migraine- 173. Fiori F, David N and Aglioti SM. Processing of pro-
associated dizziness. Am J Otol 1997; 18: 350–354. prioceptive and vestibular body signals and self-trans-
156. Fotuhi M, Glaun B, Quan SY, et al. Vestibular cendence in Ashtanga yoga practitioners. Front Hum
migraine: A critical review of treatment trials. J Neurol Neurosci 2014; 8: 734.
2009; 256: 711–716. 174. Nigmatullina Y, Hellyer PJ, Nachev P, et al. The neuro-
157. Liu F, Ma T, Che X, et al. The efficacy of venlafaxine, anatomical correlates of training-related perceptuo-
flunarizine, and valproic acid in the prophylaxis of ves- reflex uncoupling in dancers. Cereb Cortex 2015; 25:
tibular migraine. Front Neurol 2017; 8: 524. 554–562.
158. Salviz M, Yuce T, Acar H, et al. Propranolol and ven- 175. Jacobson GP and Newman CW. The development of
lafaxine for vestibular migraine prophylaxis: A rando- the Dizziness Handicap Inventory. Arch Otolaryngol
mized controlled trial. Laryngoscope 2016; 126: 169–174. Head Neck Surg 1990; 116: 424–427.

You might also like