Menière's Disease

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Handbook of Clinical Neurology, Vol.

137 (3rd series)


Neuro-Otology
J.M. Furman and T. Lempert, Editors
http://dx.doi.org/10.1016/B978-0-444-63437-5.00019-4
© 2016 Elsevier B.V. All rights reserved

Chapter 19

Menière’s disease
J.M. ESPINOSA-SANCHEZ1,2 AND J.A. LOPEZ-ESCAMEZ1,3*
1
Otology and Neurotology Group, Department of Genomic Medicine, Centre for Genomics and Oncological Research (GENYO),
Pfizer-University of Granada-Junta de Andalucia, Granada, Spain
2
Department of Otolaryngology, Hospital San Agustin, Linares, Jaen, Spain
3
Department of Otolaryngology, Complejo Hospitalario Universitario de Granada, Granada, Spain

Abstract
Menière’s disease (MD) is a chronic multifactorial disorder of the inner ear characterized by episodic ves-
tibular symptoms associated with sensorineural hearing loss, tinnitus, and aural pressure. Epidemiologic
and genomic evidence supports a genetic susceptibility with multiple biochemical pathways involved,
including the endocrine system, innate immune response, and autonomic nervous system. Allergens, infec-
tious agents, vascular events, or genetic factors could modify inner-ear homeostasis and trigger MD. The
diagnosis of MD is based on clinical criteria and requires the observation of an episodic vertigo syndrome
associated with low- to medium-frequency sensorineural hearing loss and fluctuating aural symptoms
(hearing loss, tinnitus, and/or fullness) in the affected ear. Headache is also found during the attacks
and bilateral involvement is found in 25–40% of cases. Audiologic and vestibular assessment is recom-
mended to monitor the clinical course. The treatment of MD is symptomatic to obtain relief of vestibular
episodes and preventive to limit hearing loss progression. Treatment options include sodium restriction,
betahistine, intratympanic gentamicin, or steroids and eventually surgery, such as cochlear implantation.

DEFINITION
Surgery (AAO-HNS), and the Korean Balance Society.
Menière’s syndrome is a chronic inner-ear disorder char- The classification includes two categories: definite MD
acterized by recurrent episodes of spontaneous vertigo and probable MD (Table 19.1). The diagnosis of def-
and fluctuating unilateral sensorineural hearing loss inite MD is based on clinical criteria and requires
(SNHL), tinnitus, and aural fullness. When this set of the observation of an episodic vertigo syndrome asso-
symptoms cannot be attributed to a specifically identified ciated with low- to medium-frequency SNHL and fluc-
cause, the syndrome is considered idiopathic and then it tuating aural symptoms (hearing loss, tinnitus, and/or
is referred to as Menière’s disease (MD). fullness) in the affected ear. The episodes of vertigo
The diagnostic criteria for MD have been jointly have a duration of between 20 minutes and 12 hours.
formulated by the Classification Committee of the Probable MD is a broader concept, defined by episodic
Bárány Society, the Japan Society for Equilibrium vestibular symptoms (vertigo or dizziness) associated
Research, the European Academy of Otology and Neu- with fluctuating aural symptoms occurring in a period
rotology (EAONO), the Equilibrium Committee of the from 20 minutes to 24 hours (Lopez-Escamez
American Academy of Otolaryngology-Head and Neck et al., 2015).

*Correspondence to: Dr. Jose A. Lopez-Escamez, Otology & Neurotology Group CTS495, Department of Genomic
Medicine, GENYO, Centre for Genomics and Oncological Research, Pfizer/Universidad de Granada/Junta de Andalucía,
PTS, Avda de la Ilustración, 114, 18016 Granada, Spain. Tel: +34-958-715-500-160, Fax: +34-958-63-70-71, E-mail:
antonio.lopezescamez@genyo.es
258 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
Table 19.1 patients with bilateral MD involvement increases with
Diagnostic criteria for Menière’s disease duration of illness (Vrabec et al., 2007; Huppert et al.,
2010). Bilateral MD has been reported in 25–40% of
Definite Menière’s disease affected individuals (House et al., 2006). Bilateral MD
A. Two or more spontaneous episodes of vertigo, each lasting is associated with increased vestibular symptoms and
20 minutes to 12 hours has a negative impact on health-related quality of life
B. Audiometrically documented low- to medium-frequency
(Lopez-Escamez et al., 2009).
sensorineural hearing loss in the affected ear on at least one
MD patients suffer from other disorders that are con-
occasion before, during, or after one of the episodes of
vertigo sidered MD comorbidities. These include allergic and
C. Fluctuating aural symptoms (hearing, tinnitus, or fullness) autoimmune disorders such as rheumatoid arthritis, sys-
in the affected ear temic lupus erythematosus, and ankylosing spondilytis
D. Not better accounted for any other vestibular diagnosis (Derebery and Berliner, 2010; Derebery, 2011;
Probable Menière’s disease Gazquez et al., 2011; Tyrrell et al., 2014). Several authors
A. Two or more spontaneous episodes of vertigo, each lasting have found a high prevalence of migraine in patients with
20 minutes to 12 hours MD as compared to age- and sex-matched controls
B. Fluctuating aural symptoms (hearing, tinnitus, or fullness) (Radtke et al., 2002; Shin et al., 2013). It seems
in the affected ear that migraine is more common in patients with MD
C. Not better accounted for any other vestibular diagnosis
whose family history includes episodic vertigo (Cha
et al., 2007).
Reprinted from Lopez-Escamez JA, Carey J, Chung WH, et al. (2015). When at least one first- or second-degree relative fulfills
Diagnostic criteria for Menière’s disease. J Vestib Res 25:1–7 with
criteria for MD, familial MD (FMD) is diagnosed. Family
permission from IOS Press. Available at IOS Press through http://
dx.doi.org/10.3233/VES-150549. history is observed in 5–15% of sporadic cases in popula-
tions of European descent (Morrison et al., 2009; Vrabec,
2010; Hietikko et al., 2013; Requena et al., 2014). An
autosomal-dominant pattern of inheritance with incom-
EPIDEMIOLOGY
plete penetrance is commonly found, although genetic het-
Although MD is considered the third most common erogeneity has been described in FMD (Requena et al.,
cause of vertigo after benign paroxysmal positional ver- 2014). A family history of migraine or SNHL, isolated
tigo and vestibular migraine (VM), it is a rare disease in or as a part of an episodic vestibular syndrome, is also
the general population. The occurrence varies broadly observed in some cases, suggesting an incomplete pheno-
according to the applied diagnosis criteria, source of data, type (Requena et al., 2014).
and geographic and ethnic variables.
Depending on the studies, the incidence of MD ranges
ETIOLOGYAND PATHOPHYSIOLOGY
from 8.2 to 157 per 100 000 individuals per year
(Cawthorne and Hewlett, 1954; Stahle et al., 1978; Human temporal bone studies have linked MD symp-
Celestino and Ralli, 1991). The prevalence of MD ranges toms to the accumulation of endolymph within the
from 3.5 per 100 000 inhabitants in Japan to 513 per cochlear duct (scala media) and the sacculus in the inner
100 000 in Finland (Okafor, 1984; Wladislavosky- ear. It is believed that this endolymphatic hydrops (EH)
Waserman et al., 1984; Watanabe et al., 1995; Kotim€aki begins with a disturbance of the ionic composition of the
et al., 1999; Havia et al., 2005; Harris and Alexander, scala media. However, current data support the hypoth-
2010; Tyrrell et al., 2014). MD is more common in Cau- esis that EH is an epiphenomenon associated with a vari-
casians than in other populations such as Japanese or ety of inner-ear disorders (Merchant et al., 2005), and that
Native Americans (Wiet, 1979; Ohmen et al., 2013). genetics and environmental factors contribute to its
Children are rarely affected since the disease com- development (Fig. 19.1). So, food or respiratory aller-
monly begins in the fourth or fifth decade of life, and gens, infectious agents, vascular events, or genetic factors
the prevalence increases with increasing age (Stahle could trigger an imbalance in inner-ear homeostasis.
et al., 1991; Lee et al., 1995; Tokumasu et al., 1996; Several regulatory factors, such as the innate immune
Ballester et al., 2002; Havia et al., 2005; Shojaku response, the endocrine system, or the autonomic ner-
et al., 2005; Harris and Alexander, 2010). vous system, may also influence the development of
There seems to be a slight female predominance, the partial or complete phenotype observed in familial
although the female:male ratio varies broadly among MD (Requena et al., 2014). The cumulative effect of
series (Stahle et al., 1991; Lee et al., 1995; Havia one or several triggers and the individual response
et al., 2005; Shojaku et al., 2005). No difference exists may explain the clinical heterogeneity observed in MD
in the ratio of right to left ear. Also the proportion of phenotype.
MENIÈRE’S DISEASE 259
Trigger factors Regulatory factors Clinical phenotype

Innate immune response


Endocrine response
Endolymphatic hydrops
Allergens

Aural fullness

Infectious agents
Tinnitus
Inner ear
homeostasis Sensorineural
Vascular events hearing loss

Episodic vestibular
syndrome
Genetic variants

Episodic headache
Autonomous nervous
system

Fig. 19.1. Core hypotheses for Menière’s disease. Multiple genetic or environmental factors could challenge inner-ear homeo-
stasis and trigger a partial or a complete clinical phenotype, depending on the individual susceptibility, according to several reg-
ulatory factors such as the inmate immune response, the endocrine system, and the autonomic nervous system. (Modified after
Merchant et al., 2005.)

Epidemiologic and genomic evidence supports each with MD (Kim et al., 2014), and the association of allelic
of the three major hypotheses regarding etiology of variants of MICA, TLR10, and NFKB1 genes with hear-
MD: allergy, autoimmunity, and genetic factors. Further- ing loss progression in patients with MD (Gazquez et al.,
more, one or more suspected environmental factors may 2012; Requena et al., 2013; Cabrera et al., 2014).
trigger the disease. Although preliminary candidate gene studies found
that bilateral MD was associated with allelic variants
Allergy of human leukocyte antigen (HLA) genes (Lopez-
Escamez et al., 2002) or PTPN22 (Lopez-Escamez
Among cross-sectional surveys, the prevalence of diag-
et al., 2010), these findings have not been formally rep-
nosed allergy was three times higher in those with a his-
licated. Different studies suggest the role of innate
tory of MD compared with the general population
immune response in the hearing outcome of autoimmune
(Derebery, 2000). Among patients with MD, 58% had
inner-ear disease (Pathak et al., 2011; Svrakic et al.,
a history of allergy and 41% had a positive skinprick test
2012) and MD (Requena et al., 2013; Cabrera et al.,
(Derebery, 2000), and 43% of patients had elevated
2014; Kim et al., 2014). Several mechanisms may
levels of immunoglobulin E compared with controls
explain the development of an immune-mediated
(Keles et al., 2004). Derebery and Berliner (2010) pro-
inner-ear disease: (1) cross-reactions with a cross-
posed that allergic inflammation affects the endolym-
reactive epitope (antibodies cause accidental inner-ear
phatic sac in patients with MD. Although the evidence
damage because the ear shares epitopes with a potentially
of a causal association between allergy and MD is not
harmful substance, virus, or bacterium) as suspected for
demonstrated, a food or seasonal allergy should be con-
some inflammatory diseases (Platt et al., 2013); (2) dam-
sidered as a trigger of MD and treated (Weinreich and
age to the inner ear caused by proinflammatory cytokines
Agrawal, 2014).
such as interleukin-1b (Pathak et al., 2011; Zhao et al.,
2013) or tumor necrosis factor (Svrakic et al., 2012),
Autoimmunity
as in some autoimmune diseases; or (3) inappropriate
Autoimmune inner-ear disease and MD may have an immune response or intolerance to harmless unrecog-
overlapping phenotype and several immune mechanisms nized substances combined with genetic factors that
are probably involved in the pathophysiology of MD. modify the immune response as in allergies (Greco
Some evidence supports this hypothesis, including the et al., 2012). These findings, together with the elevated
response to steroid therapy, the finding of elevated levels prevalence of several autoimmune diseases, point to
of autoantibodies or circulating immune complexes autoimmunity in MD (Gazquez et al., 2011; Tyrrell
against inner-ear antigens in the serum of some patients et al., 2014).
260 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
Genetic bases of MD 2001; Morales-Angulo and Gallo-Terán, 2005; Perez-
Fernandez et al., 2010).
Several lines of evidence support a genetic susceptibility
Hearing loss is associated with vertigo attacks in 77%
for MD. First, familial clustering was observed in MD in
of patients (Lopez-Escamez et al., 2014). It is fluctuating
Finland and Spain, and the sibling recurrence risk ratio is
in the first years, in the sense that it is episodic and revers-
45 times higher compared to the general population
ible after the crisis has subsided. Nevertheless, as the dis-
(Hietikko et al., 2013; Requena et al., 2014). Although
ease progresses, hearing worsens with each crisis and it
linkage studies were performed in several multicase fam-
does not return to the previous level. Eventually, deaf-
ilies with MD (Klar et al., 2006; Gabriková et al., 2010;
ness becomes permanent and no longer fluctuates. Ler-
Arweiler-Harbeck et al., 2011) and a locus at 12p12.3
moyez’s syndrome is a rare phenomenon in some
was defined in Swedish families (Gabriková et al.,
patients with MD. It consists of a transient improvement
2010), no single gene was associated with familial
of hearing during the onset of a vertigo attack. Tinnitus
MD. Remarkably, Requena et al. (2015) have identified
may also improve.
the first two disease-causing mutations in FAM136A and
Some patients report a previous history of hearing
DTNA genes by whole-exome sequencing. These muta-
loss, often since childhood, preceding the onset of the
tions segregated MD phenotype in a family with an
episodes of vertigo. This variant is named delayed MD
autosomal-dominant inheritance pattern in three affected
(Lopez-Escamez et al., 2015).
women in consecutive generations showing anticipation.
Tinnitus may be the initial symptom of MD, preced-
Most cases of FMD have an autosomal-dominant pattern
ing the full picture by months. It is commonly described
of inheritance, but genetic heterogeneity is observed with
as low-pitched, as a harsh, roaring, machine-like sound
suspected mitochondrial and recessive pattern (Requena
or a hollow seashell sound. At the onset of the disease,
et al., 2014). A second line of evidence for a genetic pre-
tinnitus is intermittent and appears during the attacks
disposition to MD comes from candidate gene studies.
in 83% of patients and disappears afterward (Lopez-
Although no consistent association has been replicated
Escamez et al., 2014). As the attacks recur, tinnitus also
(Gazquez and Lopez-Escamez, 2011; Hietikko et al.,
becomes permanent between episodes, although its
2012), allelic variants in MICA (Gazquez et al., 2012),
intensity may increase or the tone may change before
TLR10 (Requena et al., 2013), and NFKB1 genes
or during the episodes. Typically, patients mention that
(Cabrera et al., 2014) influence hearing loss progression
this change in their tinnitus warns them the attack is com-
in MD. Finally, the clear predilection of the disease for
ing. At the late stages, when the episodes of vertigo have
Caucasians over other ethnicities also supports a genetic
disappeared, tinnitus becomes a prominent symptom that
component (Ohmen et al., 2013).
may cause a significant impairment in quality of life.
Aural fullness is variable and more than 20% of
patients never experience it (Levo et al., 2014; Lopez-
CLINICAL MANIFESTATIONS
Escamez et al., 2014). The sensation is described as a
Numerous factors have been alleged to precipitate an feeling of pressure in the ear similar to that observed
MD attack, including stress, sleep deprivation, some when descending to land in an airplane. It commonly dis-
food, allergens, barometric pressure change, and hor- appears as the disease progresses. Hyperacusis and dipla-
monal changes (menses) (Andrews and Honrubia, 2010). cusis are also reported in patients with MD.
The occurrence of recurring episodes of spontaneous Brantberg and Baloh (2011) found that 68% of
vertigo is the main feature of MD and it is present in patients with MD described two or more of the three
96.2% of patients (Paparella and Mancini, 1985). Vertigo characteristic auditory symptoms (unilateral hearing
is the most disabling symptom, commonly described as loss, unilateral tinnitus, and unilateral aural fullness) dur-
spinning, exacerbated by head movements, and accom- ing at least half of the vertigo spells.
panied by nausea, vomiting, and sweating. Spells of Headache occurs in 41% of Menière’s attacks and
vertigo last several hours, and when they subside migraine-type headache occurs in 8% of patients during
patients complain of unsteadiness for several days. These episodes of vertigo (Lopez-Escamez et al., 2014). How-
spells are often preceded by tinnitus, aural fullness, ever, headache is not considered a diagnostic criterion
and a decrease in hearing in the affected ear. Some for MD.
patients report sudden falls with no previous warning Disease begins with the three classic symptoms in
or provocative factor, and without vertigo, loss of con- only 40% of patients (Belinchon et al., 2012). It is not
sciousness, or other neurologic symptoms. These epi- uncommon that recurrent episodes of vertigo without
sodes are named otolithic crises of Tumarkin. The auditory symptoms, or fluctuating hearing loss with or
occurrence of these vestibular drop attacks is variable, without tinnitus precede the characteristic triad of recur-
although some series report up to 72% (Kentala et al., rent vertigo, hearing loss, and ipsilateral tinnitus by
MENIÈRE’S DISEASE 261
months. The time delay between hearing loss and vertigo Table 19.2
may be more than 5 years in 20% of MD patients Menière’s disease diagnostic scale
(Pyykk€ o et al., 2013). However, most patients develop
the full picture within the first year. Certain Menière’s disease
The number of episodes of vertigo decreases during Definitive Menière’s disease plus histopathologic confirmation
follow-up, and about 70% of patients who do not present Definite Menière’s disease
Two or more definitive spontaneous episodes of vertigo lasting
with an episode of vertigo for 1 year will continue to be
20 minutes or longer
free of episodes during the following year (Perez-
Audiometrically documented hearing loss on at least one
Garrigues et al., 2008). The frequency of episodes of occasion
vertigo shows a rapid decline over the first 8–9 years, Tinnitus or aural fullness in the treated ear
and subsequently stabilizes over the following 10 years Other causes excluded
before declining gradually (Green et al., 1991; Stahle Probable Menière’s disease
et al., 1991; Perez-Garrigues et al., 2008). One definitive episode of vertigo
The presence of vestibular abnormalities on bedside Audiometrically documented hearing loss on at least one
examination is well recognized in MD. Head-shaking occasion
nystagmus and vibration-induced nystagmus are com- Tinnitus or aural fullness in the treated ear
mon vestibular signs in patients with MD (Marques Other causes excluded
Possible Menière’s disease
and Perez-Fernandez, 2012; Neff et al., 2012). Three
Episodic vertigo of the Menière type without documented
or more beats of spontaneous nystagmus or the presence
hearing loss, or sensorineural hearing loss fluctuating or
of nystagmus for at least 5 seconds after vigorous head fixed, with disequilibrium but without definitive episodes
shaking is considered as abnormal. Spontaneous nystag- Other causes excluded
mus is often absent between relapses, although a
horizontal-torsional nystagmus may be observed during Reproduced from Committee on Hearing and Equilibrium (1995).
an attack. Classically it has been described as a first irri-
tating phase with nystagmus beating toward the affected The Bárány Society’s criteria are more accurate, since
ear, a second paretic phase with the nystagmus beating they specify the duration of the episodes of vertigo (from
toward the opposite ear, and a final recovery phase in 20 minutes to 12 hours) and the type of hearing loss (low-
which again nystagmus beats toward the pathologic to medium-frequency SNHL) and require fluctuating
ear. Thus, nystagmus direction varies, beating either auditory/aural symptoms. The previous 1995 AAO-
toward the affected ear or the healthy ear, so it cannot HNS criteria set up four categories (certain, definite,
be considered a localizing finding. Positional nystagmus probable, and possible), whereas the Bárány Society’s
can be found either during or between attacks, although criteria only consider two categories: definite and
the frequency varies widely among different series probable MD.
(Hulshof and Baarsma, 1981; Dobie et al., 1982). The
head impulse test (HIT) yields positive results in less than
FUNCTIONAL TESTS
half of MD patients.
Vestibulospinal function tests are consistent with a Audiologic evaluation
peripheral vestibular syndrome: falling or deviation
PURE-TONE AUDIOMETRY
toward the side of the lesion is commonly observed in
the Romberg test with eyes closed and during tandem The AAO-HNS established a hearing staging system,
walking. according to the pure-tone thresholds at 0.5, 1, 2, and
3 kHz obtained in the audiogram (Table 19.3). Audiome-
trically documented fluctuating low-tone unilateral
DIAGNOSIS
SNHL is the key to the diagnosis of MD when facing
The diagnosis of MD is based on clinical criteria, since patients with an episodic vestibular syndrome. With
there is no biologic marker. The traditional criteria were follow-up, it is easy to document fluctuation when recov-
established by the Committee on Hearing and ery is appreciated, thus supporting the diagnosis of MD.
Equilibrium of the AAO-HNS (1995), and have been A shift in pure-tone thresholds for bone conduction by at
widely used for the last 20 years (Table 19.2). The current least 30 dB hearing level at each of two adjacent frequen-
diagnostic criteria have been developed by consensus cies below 2000 Hz is required for unilateral MD. The
among the Bárány Society, the Japan Society for Equilib- low frequencies (250 and 500 Hz) are typically affected
rium Research, the EAONO, the AAO-HNS, and the at the earlier stages. As disease progresses, all frequen-
Korean Balance Society as part of the International Clas- cies may be involved and the audiogram pattern flattened
sification of Vestibular Disorders (Table 19.1). at a moderate or severe level (Belinchon et al., 2011).
262 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
Table 19.3 Vestibular testing
Staging of definite and certain Menière’s disease CALORIC TESTS
Stage Pure-tone average (dB) Bithermal caloric irrigation with computerized electro-
nystagmography or videonystagmography has been the
1 25 main laboratory test to evaluate vestibulo-ocular reflex
2 26–40 (VOR) function. Caloric tests assess horizontal semicir-
3 41–70 cular canal function, with the percentage of unilateral
4 >70 caloric weakness or canal paresis as the main outcome
Reproduced from Committee on Hearing and Equilibrium (1995).
measure. Unilateral vestibular hypofunction on caloric
Staging is based on the four-tone average (arithmetic mean rounded to testing is observed in up to 75% of unilateral MD patients
the nearest whole number) of the pure-tone thresholds at 0.5, 1, 2, and (Wang et al., 2012), although it is worth noting that a nor-
3 kHz of the worst audiogram during the interval 6 months before treat- mal bithermal caloric response has been reported in up to
ment. This is the same audiogram that is used as the baseline evaluation 50% of patients in some series. Unilateral canal paresis
to determine hearing outcome from treatment. Staging should be
applied only to cases of definite or certain Menière’s disease.
usually indicates the involved ear, but it has also been
demonstrated in 19% of patients on the normal side
(Proctor, 2000).
ELECTROCOCHLEOGRAPHY (ECOG)
ECoG is a neurophysiologic technique in which an audi-
VIDEO-HEAD IMPULSE TEST (VHIT)
tory evoked potential is obtained in response to brief
sound stimuli and recorded by an intratympanic or extra- This is a video-oculography device that allows assess-
tympanic (noninvasive) electrode. The cochlear micro- ment of the VOR at high frequencies during HIT. The
phonic and the summating potential (SP) are generated system is becoming a bedside technique as it is noninva-
by the hair cells of the organ of Corti, whereas the sive, fast, and more accessible than the scleral search coil
compound action potential (AP) of the auditory nerve (MacDougall et al., 2009). The vHIT not only records
represents the summed synchronized response of many refixation saccades but also demonstrates early saccades,
individual nerve fibers. Testing parameters include laten- invisible to the human eye (covert saccades). The equip-
cies and amplitudes of SP and AP, and SP/AP amplitude ment may provide an objective measurement of VOR
ratio, and area under the curve of SP/AP ratio. gain when head impulses are performed in the plane of
Changes in the SP response can reflect pressure differ- each of the six semicircular canals.
ences between the scala media and the scala vestibuli, It has been reported that 67% of patients with MD
indicating excessive fluid pressure, thus deforming the show a reduced VOR gain in at least one semicircular
basilar membrane toward the scala tympani, so that canal when the six canals are tested; the posterior semi-
enhanced-amplitude SP is thought to reflect EH. SP/AP circular canal of the affected ear is the most frequently
ratio is the most common parameter for diagnosis of EH. involved canal (Zulueta-Santos et al., 2014).
However, normal ECoG responses have also been
reported in patients with EH.
VESTIBULAR-EVOKED MYOGENIC
Increases in SP amplitude with an enlarged SP/AP
POTENTIALS (VEMPS)
ratio and a prolonged AP latency shift have been observed
in patients with MD (Ge and Shea, 2002; Ferraro These are otolith-mediated, middle-latency reflexes that
and Durrant, 2006). Nevertheless, the sensitivity and are recorded from sternocleidomastoid (cVEMPs) or
specificity of SP/AP ratio for detecting MD are highly infraocular (oVEMPs) electromyography in response
variable, with a low sensitivity and higher specificity to high-intensity auditory stimuli (air conduction) or
(Lamounier et al., 2014). The use of tone bursts instead high-frequency vibratory stimulation (bone conduction).
of clicks, and the combination of SP/AP ratio with the area Air conduction is preferred for cVEMPs, while bone-
under the curve SP/AP may increase the sensitivity of conducted vibration is mostly used in oVEMPs. VEMPs
ECoG (Al-Momani et al., 2009; Claes et al., 2011). It show a biphasic waveform with a positive and a negative
has also been reported that the sensitivity of ECoG peak. The short-onset latency of the cVEMPs is gener-
increases with the duration and severity of disease ated by primary vestibular afferents projecting to the
(Kim et al., 2005; Takeda and Kakigi, 2010). When hear- vestibular nuclei and hence via the ipsilateral medial ves-
ing thresholds reach 60 dB, EcoG cannot be used. ECoG tibulospinal tract to the accessory nucleus. It is widely
has been performed for determining hearing outcome accepted that cVEMPs evaluate the integrity of the sac-
(Moon et al., 2012) and to monitor the response to intra- culus and the inferior vestibular nerve, whereas oVEMPs
tympanic steroid therapy (Martin-Sanz et al., 2013a). primarily evaluate contralateral utriculus and superior
MENIÈRE’S DISEASE 263
vestibular nerve. The response parameters commonly DIFFERENTIAL DIAGNOSIS
used are latencies and interpeak amplitude of the
Table 19.4 includes the major causes of episodic vestib-
response. VEMPs are currently a standardized technique
ular syndromes. When facing a patient with recurrent
and provide a quick and noninvasive method of assessing
vertigo and migraine, the biggest challenge is to distin-
otolith function in patients with an episodic vestibular
guish between VM and MD (Espinosa-Sanchez et al.,
syndrome.
2014b). Headache is a common complaint in 70% of
Patients with unilateral MD usually show abnormali-
patients with MD (Eklund, 1999), and migraine is a
ties in VEMPs with reduced or absent responses,
comorbidity in over 30% of MD patients (Parker,
although at the initial stage an augmented response is
1995; Shin et al., 2013). Further, it has also been reported
sometimes registered (Young et al., 2002; Huang et al.,
that 45% of patients with MD experienced at least one
2011; Manzari et al., 2011; Murofushi et al., 2011;
migraine symptom during the vertigo episode (Radtke
Taylor et al., 2011; Winters et al., 2011; Young, 2013;
et al., 2002). During vertigo spells, patients with MD
Jerin et al., 2014). It has also been reported that the asym-
experienced headache in 41% and migraine-type head-
metry ratio of cVEMPs increases as the disease pro-
ache in 8.4% (Lopez-Escamez et al., 2014).
gresses (Young et al., 2003). Nevertheless, the
Current criteria for MD and VM (Lempert et al., 2012;
sensitivity and specificity of VEMPs in diagnosing
Lopez-Escamez et al., 2015) provide useful tools for
MD are as low as 50% and 49%, respectively.
establishing a diagnosis, but the clinical pictures of both
diseases often overlap, especially at the beginning of the
IMAGING TECHNIQUES disease. These criteria require other causes to be
excluded, although when patients have two different
Computed tomography (CT) images reveal that the ves-
types of attacks, one fulfilling VM criteria and the other
tibular aqueduct is significantly shorter and narrower and
MD, both disorders should be diagnosed.
has a smaller external aperture on average in patients
The association of unilateral auditory symptoms is the
with MD, both in the affected and contralateral ear
most useful clinical differential characteristic to distin-
(Krombach et al., 2005; Miyashita et al., 2012).
guish between MD and VM (Brantberg and Baloh,
Although these findings can contribute to explain the
2011; Lopez-Escamez et al., 2014). Nevertheless, audi-
pathogenesis of the disease, their diagnostic significance
tory symptoms in VM increase with follow-up, in such
is limited.
a way that hearing loss appears in 38% of patients with
Magnetic resonance imaging (MRI) obtained after
VM after a median follow-up of 9 years (Radtke
intratympanic or intravenous administration of gadolin-
et al., 2012).
ium has allowed not only in vivo visualization of the
SNHL is uncommon in VM; when it is present, audi-
membranous labyrinth (Naganawa et al., 2006), but also
ometry usually demonstrates a low-frequency, mild to
the demonstration of EH in humans diagnosed of MD
moderate, bilateral SNHL, usually episodic, and it
(Nakashima et al., 2007; Naganawa et al., 2008). Various
authors have shown EH in 90% or more of patients with
definite MD when specific inner-ear MRI protocols were Table 19.4
performed (Fiorino et al., 2011; Pyykk€ o et al., 2013). Differential diagnosis of Menière’s disease
Heavily T2-weighted three-dimensional fluid-attenuated
Autosomal-dominant sensorineural hearing loss type 9
inversion recovery sequences on 3-T scanner appear to
(DFNA9) caused by COCH gene
offer the best images. In particular, as gadolinium reaches Autoimmune inner-ear disease
the perilymphatic space a signal void appears, corre- Cerebrovascular disease (transient ischemic attack/infarction/
sponding to the distended endolympathic space. hemorrhage in the vertebrobasilar system)
Several studies have found a good correlation Cogan’s syndrome
between cochlear hydrops on MRI and abnormal ECoG Endolymphatic sac tumor
(Seo et al., 2013) or abnormal VEMP (Katayama et al., Meningiomas and other masses of the cerebellopontine angle
2010; Fiorino et al., 2011). Nevertheless, the extent of Neuroborreliosis
EH visualized on MRI does not always correlate with Otosyphilis
the severity of cochleovestibular symptoms. MRI is Susac syndrome
emerging as a useful tool not only for diagnosis of Third-window syndromes (perilymph fistula, canal dehiscence,
enlarged vestibular aqueduct)
EH, but also for early detection of contralateral involve-
Vestibular migraine
ment, to evaluate the permeability of the round and oval Vestibular paroxysmia (neurovascular compression syndrome)
windows to intratympanic drugs and to document pro- Vestibular schwannoma
gression of the disease (Miller and Bykowski, 2014; Vogt–Koyanagi–Harada syndrome
Gu et al., 2015).
264 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
progresses much more slowly than in MD. Fluctuating Table 19.5
SNHL is not specific for MD, as it has also been observed Functional level scale
in VM and other disorders such as autoimmune inner-ear
disease, Cogan’s syndrome, otosyphilis, and enlarged Regarding your current state of overall functioning, not just
vestibular aqueduct syndrome. during attacks, check the one that best applies:
Patients with MD show significantly more abnormal 1. My dizziness has no effect on my activities at all
2. When I am dizzy, I have to stop what I am doing for a while,
results than patients with VM using the head thrust test,
but it soon passes and I can resume activities. I continue to
head-shaking nystagmus, vibration-induced nystagmus,
work, drive and engage in any activity I choose without
bithermal caloric tests, rotatory chair testing, and VEMPs restriction. I have not changed any plans or activities to
(Neff et al., 2012; Wang et al., 2012; Bl€ odow et al., accommodate my dizziness
2014). Obtaining a CT scan is useful to rule out other 3. When I am dizzy, I have to stop what I am doing for a while,
causes of episodic vestibular symptoms mimicking but it does pass and I can resume activities. I continue to
MD, such as canal dehiscence or enlarged vestibular work, drive, and engage in most activities I choose, but
aqueduct. MRI is routinely requested to exclude a vestib- I have had to change some plans and make some allowance
ular schwannoma. for my dizziness
4. I am able to work, drive, travel, take care of a family, or
engage in most essential activities, but I must exert a great
PROGNOSIS deal of effort to do so. I must constantly make adjustments in
The clinical course of MD is variable, with episodes my activities and budget my energies. I am barely making it
occurring at irregular intervals. Usually as the disease 5. I am unable to work, drive, or take care of a family. I am
progresses, attacks decrease in frequency, and hearing unable to do most of the active things that I used to. Even
essential activities must be limited. I am disabled
loss becomes permanent, as does the tinnitus (Perez-
6. I have been disabled for 1 year or longer and/or I receive
Garrigues et al., 2008).
compensation (money) because of my dizziness or balance
Without treatment, 57% of patients had complete con- problem
trol of vertigo at 2 years, and 71% had complete control
after an average of 8.3 years (Silverstein et al., 1989). Reproduced from Committee on Hearing and Equilibrium (1995).
Dizziness severity, bilateral SNHL, and comorbid
migraine are associated with a decrease in health-related
quality of life (Lopez-Escamez et al., 2009; Porter and We recommend utilizing the minimum effective dosage
Boothroyd, 2015). The functional scale developed by of the drug; not to make use of vestibular suppressant for
the AAO-HNS is useful to monitor the impact of MD more than 5 days; and avoid the combination of two
on daily activities (Table 19.5). drugs with antidopaminergic effects.

TREATMENT Preventive treatment


There is no consensus about MD treatment (Sajjadi and The main goal of preventive treatment is to improve
Paparella, 2008; Harcourt et al., 2014). New guidelines patients’ quality of life. This may be achieved by reduc-
for treatment of vestibular disorders, including MD, ing the frequency, duration, and severity of vertigo spells.
are being developed by the EAONO. Preventive treatment includes lifestyle and dietary mod-
It is essential to provide information to patients about ifications, pharmacologic therapy, and in some cases sur-
the natural history of the disease, discussing the thera- gical procedures. Other treatment options, such as
peutic options and their potential adverse effects, and hearing aids, cochlear implants, and the Meniett device,
encouraging them to participate actively in making deci- are also discussed below.
sions that affect their lives. This approach maximizes
treatment compliance. Treatment of MD can be catego- Lifestyle and dietary modifications
rized as either acute or preventive.
Patients with MD are counseled to follow a regular daily
routine, and avoid triggers such as stress, barometric
Acute treatment
pressure change, fatigue, or sleep deprivation. Alcohol,
Acute attacks of MD are self-limited and often subside in coffee, and tobacco are traditionally restricted, although
a few hours. Treatment of the acute episode is merely the efficacy of these measures has not been demonstrated
symptomatic, almost always with vestibular suppres- in randomized controlled trials (Luxford et al., 2013).
sants and antiemetics. Benzodiazepines are preferred The most important dietary recommendation is a high
as a vestibular suppressant while antidopaminergic water intake and a very lowsodium diet. Several evi-
agents are useful when nausea or vomiting appears. dences support that arginine vasopressin (AVP) regulates
MENIÈRE’S DISEASE 265
water reabsorption in the inner ear by aquaporin 2 chan- vasodilation of the arterioles of the stria vascularis, and
nels, including: (1) the elevated plasma levels of AVP in also in the posterior semicircular canal ampulla. Thus,
patients with MD; (2) the accumulation of endolymph in betahistine would reduce endolymphatic pressure by
AVP-treated guinea pigs and rats; and (3) the downregu- achieving a reduction in the production of, and an incre-
lation of V2 receptors in the inner ear of AVP-treated rats ment in the re-absorption of, endolymph (Dziadziola
(Egami et al., 2013). et al., 1999; Laurikainen et al., 2000; Ihler et al., 2012).
Sodium restriction is supported by the hypothesis that Furthermore, experimental data regarding betahistine
an increase of endolymphatic pressure can lead to the also suggest a decrease in the sensory input from the ves-
rupture of membranes in the scala media. So, a high salt tibular receptors (Botta et al., 1998; Chávez et al., 2005;
intake would increase sodium plasma levels and this in Desmadryl et al., 2012). On the other hand, there is grow-
turn would raise endolymphatic levels of sodium causing ing evidence that betahistine acts also at various levels of
enlargement of endolymphatic pressure. The EH would the central nervous system. Animal and human studies
lead to distension and ruptures in Reissner membrane indicate betahistine blocks the presynaptic histamine
with extravasation of endolymph into perilymph. H3 receptors on the histaminergic nerve terminals origi-
Patients are advised to eliminate the use of salt at the nating from the tuberomammillary nuclei of the posterior
table and limit its use during cooking and baking. Prod- hypothalamus, thus increasing histamine turnover and
ucts with high-sodium content should be avoided. These release in the vestibular nuclei (Lacour, 2013). Likewise,
recommendations are based on expert opinions but evi- betahistine may participate in the mechanisms of hista-
dences are lacking. minergic modulation of glycine and gamma-
An increase in water intake is presumed to reduce the aminobutyric acid (GABA) release in the vestibular
severity of MD symptoms by decreasing the systemic nuclei that may contribute to rebalancing neural activity
AVP level (Naganuma et al., 2006). Thus, it seems after a unilateral vestibular loss (Bergquist et al., 2006).
reasonable to advise a high water intake to reduce Na + These actions on the histaminergic system may promote
concentration in serum and endolymph. Since changes and facilitate central vestibular compensation (Tighilet
in endolymphatic osmolarity may facilitate the onset of et al., 2007). Finally, experimental data suggest that
vertigo, we recommend increased water intake with a the upregulation of the histamine caused by the betahis-
low level of Na + and Ca2 + to prevent dehydration and tine would induce excitatory effects on the neuronal
reduce the number of vertigo spells. This recommenda- activity of cortical and subcortical structures. This
tion is based on the finding of elevated plasma levels arousal effect would facilitate sensorimotor and cogni-
of AVP in patients with MD (Egami et al., 2013). tive activity, necessary for recovery after vestibular func-
Since a food or inhalant allergy may provoke MD tion loss (Lin et al., 1990).
symptoms in some patients with a genetic predisposition, The effect of betahistine is dose- and duration-
some authors recommend food allergen avoidance and dependent (Tighilet et al., 2005; Ihler et al., 2012). The
even specific immunotherapy in those patients with standard dosage is 48–96 mg/day, although a higher dos-
allergy and MD (Derebery, 2011; Banks et al., 2012; age between 288 and 480 mg/day is sometimes used
Weinreich and Agrawal, 2014). Placebo-controlled trials (Strupp et al., 2008; Lezius et al., 2011). Adverse effects
are needed to confirm the benefits of these measures. are rare, mild, and self-limiting. A cutaneous hypersen-
sitivity reaction is the most frequently reported com-
PHARMACOLOGIC THERAPY plaint. Nausea, vomiting, epigastric pain, and headache
are occasionally reported, especially with higher doses
Betahistine
(Jeck-Thole and Wagner, 2006; Benecke et al., 2010).
This drug is broadly used worldwide, except for The clinical efficacy of betahistine has been evaluated
the USA, since it has not been approved by the US in several trials and there is conflicting evidence.
Food and Drug Administration. Betahistine is a structural A Cochrane review concluded that there was not enough
analog of histamine that acts as a weak partial postsynap- evidence to say whether betahistine has any effect on the
tic histamine H1 receptor agonist and presynaptic H3 frequency or duration of the episodes of vertigo MD
receptor antagonist, with no effect on postsynaptic (James and Burton, 2001). Subsequently, a meta-analysis
H2 receptors (Gbahou et al., 2010). The mechanism has been performed supporting the therapeutic benefit of
of action of the drug appears to depend mainly on its betahistine for both MD and vestibular vertigo (Nauta,
action on H3 receptors mediated by two metabolites, ami- 2014). Betahistine 144 mg/day yields similar vertigo
noethylpyridine and hydroxyethylpyridine (Bertlich control rate as intratympanic dexamethasone (Albu
et al., 2014). et al., 2015). However, a recent long-term, double-blind,
Experimental studies in animals demonstrate that randomized placebo-controlled clinical trial (BEMED
betahistine improves labyrinthine microcirculation by study), using 48 or 114 mg/day betahistine, has shown
266 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
that betahistine has no beneficial effect in MD (Adrion improve inner-ear function through ion or water transport
et al., 2016). Further long-term randomized, placebo- mechanisms influencing cochlear fluid homeostasis
controlled clinical trials with higher dosages of betahis- (Fukushima et al., 2004; Alles et al., 2006; Otake
tine are warranted to confirm the findings of the BEMED et al., 2009; Nevoux et al., 2015).
trial. Clinical research should also focus on identifying Few studies have investigated the effect of oral steroids
biologic markers or clinical predictors for betahistine in MD (Morales-Luckie et al., 2005; Fisher et al., 2012),
response in MD. and there are no randomized clinical trials showing any
benefit from oral steroids in MD in the long term. However,
Diuretics comorbid systemic autoimmune disorders, long-lasting
recurrent episodes of vertigo, or a sudden drop in hearing
Diuretics are commonly used in MD patients, especially
thresholds are situations that could be managed with high
in the USA, where they are the primary mode of therapy.
doses of oral steroids during few weeks.
Diuretics act by diminishing sodium reabsorption at dif-
Animal studies demonstrate that intratympanic delivery
ferent sites in the nephron, thereby increasing urinary
results in significantly higher inner-ear levels of steroids as
sodium and water loss. This reduction of extracellular
compared with systemic administration. Moreover, intra-
volume is supposed to decrease endolymphatic pressure
tympanic delivery avoids the well-known adverse effects
and volume, either by increased drainage of endolymph
of systemic administration: osteoporosis, diabetes melli-
or a reduction in its production at the stria vascularis.
tus, hypertension, peptic ulcer, cataracts, and endocrine
Thiazides, such as hydrochlorothiazide, are the most
disorders. Compared with gentamicin therapy, the main
frequently used diuretics in patients with MD. The side-
advantage of intratympanic corticosteroids is the absence
effects of diuretics are common. Thiazides produce
of risk of hearing loss. Furthermore, this is a low-cost
hyponatremia, hypochloremia, hypokalemia, and meta-
and safe technique. Residual tympanic perforation is the
bolic alkalosis, whereas potassium-sparing diuretics
main risk.
cause hyperkalemia. Loop diuretics, e.g., furosemide,
Experimental studies have demonstrated that, although
generate hypokalemia, hypovolemia, hypochloremic
after intratympanic injection, methylprednisolone reaches
alkalosis, dilutional hyponatremia, and hyperuricemia.
higher concentrations in endolymph and perilymph than
Significantly, loop diuretics can cause hearing loss, gen-
dexamethasone, the latter drug may be more efficacious
erally temporary, and it must be remembered that amino-
as it is absorbed more rapidly by endocytosis into the stria
glycosides act synergistically with loop diuretics to
vascularis and surrounding tissues, where it acts intracellu-
produce an unexpectedly high incidence of ototoxicity.
larly (Parnes et al., 1999; Hamid and Trune, 2008).
Acetazolamide, a carbonic anhydrase inhibitor, gives rise
Clinical studies are heterogeneous and differ in drug
to hypokalemia, hyperchloremic metabolic acidosis, and
delivery protocols, type, and concentration of steroids
urinary lithiasis.
(Boleas-Aguirre et al., 2008; Herraiz et al., 2010;
The effects of diuretics in patients with MD have been
Martin-Sanz et al., 2013b; McRackan et al., 2014a).
analyzed by the Cochrane Collaboration (Thirlwall and
High-dose dexamethasone (16 mg/mL) appears to pro-
Kundu, 2006), concluding that the effect of diuretics in
vide better outcome than a lower dosage (4 mg/mL)
MD cannot be evaluated due to lack of properly con-
(Casani et al., 2012). The only prospective randomized
ducted trials. Diuretics can be considered a second- or
double-blind trial considered for a Cochrane systematic
third-line drug that may be used alone or in combination
review found 82% of complete control of vertigo over
with betahistine when this drug fails to reduce the fre-
placebo (57%) (Garduño-Anaya et al., 2005). The
quency of vertigo spells.
authors of this review concluded that, although a single
trial provided limited evidence, intratympanic corticoste-
Steroid therapy
roids have demonstrated a statistically and clinically sig-
The use of corticosteroids in autoimmune diseases, the nificant improvement of the frequency and severity or
beneficial effect of steroids in the treatment of sudden vertigo (Phillips and Westerberg, 2011). Large prospec-
SNHL (Li et al., 2015), and the participation of inflam- tive randomized controlled clinical trials are needed. At
matory and immune mechanisms in the pathophysiology present, when first-line treatment fails to control vertigo,
of MD have led to the consideration of corticosteroids as it is a common practice to administer intratympanic cor-
a treatment option in MD (Lopez-Escamez et al., 2007, ticosteroids if the patient still has functional hearing.
2010; Hamid and Trune, 2008; Hu and Parnes, 2009).
The mechanism of action of corticosteroids in MD is
Intratympanic gentamicin therapy (IGT)
not limited to their anti-inflammatory and immunosup-
pressive effects in the cochlea, including the stria vascu- The aminoglycoside antibiotics are used in the manage-
laris. They can increase labyrinthine circulation and ment of MD at low dosage to produce a partial vestibular
MENIÈRE’S DISEASE 267
ablation. Initially, aminoglycosides were administered such as the appearance of hearing loss or clinical signs
systemically in MD, but two major drawbacks appeared: of vestibular hypofunction, including spontaneous nys-
hearing loss and bilateral vestibular hypofunction, with tagmus, head-shaking nystagmus, or a positive head
severe ataxia and oscillopsia. thrust test. For this purpose, vHIT has been shown to
The aim of IGT is to obtain a long-lasting, nonfluctu- be a valuable tool in assessing vestibulo-ocular changes
ating, peripheral vestibular hypofunction capable of after IGT (Marques et al., 2015). Recent studies have
being centrally compensated. As compared with sys- shown that “on demand” low-dose protocols yield a
temic administration, intratympanic therapy has many lower risk of hearing loss and posttreatment instability
advantages: it is an office-based procedure that avoids with good long-term vertigo control (Manrique-Huarte
toxicity in the contralateral ear or other organs and yields et al., 2011; Casani et al., 2014; Quaglieri et al., 2014).
a higher concentration of drug in the inner ear. This variety of schedules and protocols makes it diffi-
The mechanism of vestibular toxicity of gentamicin is cult to compare results among different studies (Cohen-
not well understood. Animal models suggest that ototox- Kerem et al., 2004). Based on the results of only two studies
icity is caused by hair cell apoptosis induced by overpro- that fulfilled the methodologic inclusion criteria, a
duction of reactive oxygen species, including oxygen Cochrane review concluded that intratympanic gentamicin
ions, free radicals, and peroxide (Choung et al., 2009). seems to be an effective treatment for vertigo complaints in
There is also increasing evidence that programmed cell MD, although it highlights the risk of hearing loss (Pullens
death is a consequence of aminoglycoside interference and van Benthem, 2011). A meta-analysis pooling pub-
with mitochondrial protein synthesis, stimulation of lished data has demonstrated an 87.5% substantial vertigo
N-methyl-D-aspartate receptors, and activation of the control rate with significant tinnitus improvement, and
caspase pathway. minimal hearing deterioration (Huon et al., 2012).
Histologic studies show that, after IGT, there is atro- Recently, a retrospective study supports the use of intra-
phy of the neuroepithelium of the semicircular canal cris- tympanic gentamicin in patients with Tumarkin attacks
tae ampullaris and fibrosis and edema of the stroma, (Viana et al., 2014). Remarkably, IGT can achieve better
whereas the utricular macula is relatively spared. IGT control of vertigo than corticosteroid injection without sig-
causes greater loss of type I than type II vestibular hair nificant differences in final hearing level (Casani et al.,
cells because they concentrate more aminoglycoside 2012; Gabra and Saliba, 2013). When IGT fails to control
(Lyford-Pike et al., 2007). Damage to the vestibular dark vertigo, a reasonable option is exploratory tympanotomy
cells also has been observed. With regard to the cochlea, and direct application of gentamicin over the round win-
the basal turn is the region most susceptible to permanent dow and the oval-window niches (Rah et al., 2015).
loss of hair cells and replacement by supporting cells.
Outer hair cells are more damaged than inner hair cells, Other drugs
resulting in high-frequency hearing loss.
Vasodilators have been used frequently in the treatment of
Gentamicin can be delivered to the inner ear using dif-
MD because they are supposed to improve microcircula-
ferent application methods (Salt and Plontke, 2009;
tion of the labyrinth, thus mitigating hypothetic ischemia.
McCall et al., 2010; Miller and Agrawal, 2014). Usually,
It also has been proposed that vasodilators work by reduc-
IGT is used, although new techniques are been develop-
ing endolymphatic pressure or even by inhibiting vestib-
ing to deliver drugs directly into the inner ear. Once the
uwel et al., 2003).
ular nuclei activity (Smith et al., 2002; D€
agent is in the middle-ear cavity it diffuses mainly
Although their efficacy is questionable, and their mecha-
through the round-window membrane, although it may
nism of action is uncertain, calcium channel blockers are
also enter through the oval-window membrane (King
among the most widely prescribed drugs for the manage-
et al., 2013). However, clearance via the eustachian tube
ment of MD (Tapia-Toca et al., 2009).
leads to a variable amount of substance reaching inner-
Extract of ginkgo biloba is prescribed for a wide vari-
ear fluids.
ety of disturbances, including tinnitus and dizziness.
Treatment schedules for IGT can be categorized as
Several mechanisms of action have been proposed but
fixed-dose protocols, instillations on request, and titra-
there is no convincing evidence from trials of sufficient
tion techniques. The dose is preset in the fixed-dose pro-
methodologic quality to support the use of ginkgo biloba
tocols; these protocols differ depending on the amount of
extract for patients with MD (Espinosa-Sanchez
drug delivered and the frequency and number of doses;
et al., 2014a).
dosing may be daily, multiple daily, weekly, monthly,
or continuous. When instillations are performed on
SURGICAL TREATMENT
request, a low dose is given, which is repeated only if ver-
tigo spells persist or recur. In the titration technique, gen- Surgical management of MD is indicated in patients
tamicin is administered until a certain goal is achieved, when medical and intratympanic therapies fail to control
268 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
vertigo, and represents less than 5% of patients. For sev- retrolabyrinthine, and retrosigmoid approach. Transla-
eral reasons, surgery should be carefully considered only byrinthine vestibular neurectomy sacrifices hearing
in certain patients. First, although the natural history of whereas with the other three approaches it is possible
MD is variable, as the disease progresses the frequency to preserve hearing. The need for a craniotomy involves
of vertigo spells usually decreases but hearing loss is fur- the risks of a cerebrospinal fluid leak, brain edema, men-
ther aggravated. Secondly, the risk of bilaterality ranges ingitis, and intracranial bleeding. Middle fossa neurect-
from 25% to 40% and these patients have chronic dis- omy presents a major risk of facial nerve damage
equilibrium and severe bilateral SNHL. Since there is compared with the posterior approach. When compared
no biologic marker for bilateral involvement, patients with intratympanic gentamicin, vestibular neurectomy
should not undergo surgery within the first 10 years. This achieves higher vertigo control rates (Hillman et al.,
would limit surgery to a few cases with persistent vertigo 2004; Colletti et al., 2007).
after a very long follow-up. It is also remarkable that the
benefits of surgery to control Tumarkin crisis have not Labyrinthectomy
been demonstrated.
Several procedures have been described for the surgi- This is a hearing-destructive procedure reserved for
cal treatment of MD (Sismanis, 2010; Teufert and patients with nonfunctional hearing. Transmastoid labyr-
Doherty, 2010). The selection of a particular surgical inthectomy allows a complete removal of all neuroe-
technique depends on factors such as severity of disease, pithelium with a 95–99% rate of vertigo control.
hearing status, and presence of unilateral versus bilateral
disease; the status of hearing is the most important factor. Cochlear implantation
With regard to preservation of hearing, surgical proce- Cochlear implantation (CI) has been shown to be bene-
dures for MD can be broadly classified as either destruc- ficial in patients with MD who progress to bilateral
tive or nondestructive. Nondestructive procedures severe to profound SNHL or unilateral MD with contra-
include endolymphatic sac surgery (ESS), either shunt lateral hearing loss from another cause (Fife et al., 2014;
or decompression, selective vestibular neurectomy, and Vermeire et al., 2014; Samy et al., 2015). The hearing
semicircular canal occlusion. outcome seems similar to that in other patients who
undergo CI, although those patients with inactive MD
Endolymphatic sac surgery
show worse speech recognition scores (McRackan
ESS has been a controversial subject that has generated a et al., 2014b). Previous vestibular surgery, including
great amount of literature. It is thought that this procedure labyrinthectomy and selective vestibular nerve section,
relieves a supposed high endolymphatic pressure by does not contraindicate CI; furthermore, simultaneous
means of drainage into the mastoid or into the subarach- labyrinthectomy and CI is an increasingly frequent
noid space. Although several case series report vertigo option (Hansen et al., 2013; Martens et al., 2013;
control between 60% and 80%, a Cochrane review has MacKeith et al., 2014; Perez-Garrigues et al., 2015).
concluded that there is insufficient evidence for the ben- Some studies point out that vestibular symptoms may
eficial effect of ESS in MD (Hu and Parnes, 2010; improve after CI (McRackan et al., 2014b). CI is also
Pullens et al., 2013). A retrospective chart review has an option for tinnitus relief in patients with bilateral
found no significant differences in vertigo control classes SNHL and even single-sided deafness (Van Heyning
when IGT is compared with ESS (Paradis et al., 2013). It et al., 2008; Vermeire and Van de Heyning, 2009;
is also remarkable that a recent study suggests that ESS Ramos-Macías et al., 2015).
may prevent MD from developing in the contralateral ear
(Kitahara et al., 2014). OTHER THERAPIES
Semicircular canal occlusion Transtympanic ventilation tube insertion
Semicircular canal occlusion has also been utilized to Grommet insertion is a popular treatment option for MD
control intractable vertigo in MD. Triple plugging or in some countries (Smith et al., 2005; Park et al., 2009;
only horizontal semicircular canal occlusion has been Ogawa et al., 2015). However, the results are controver-
described, with good vertigo control rates and hearing sial and a placebo effect has been claimed as there are no
preservation (Yin et al., 2008; Charpiot et al., 2010). randomized controlled clinical trials.

Vestibular neurectomy Meniett device


Vestibular nerve section can be accomplished through Therapy with the Meniett device (Medtronic Xomed,
four approaches: middle fossa, translabyrinthine, Jacksonville, FL, USA) is based on the hypothesis that
MENIÈRE’S DISEASE 269
patients with MD experience changes in their symptoms 2012). We also discuss with the patient the therapeutic
depending on barometric pressure variations. The options and their potential adverse effects. Patients have
Meniett device is a portable system that applies positive to express their preferences and we encourage them to
low-pressure pulses to the external ear canal. A close- actively participate in making decisions that affect
fitting ear cuff connects the device’s tubing to the exter- their lives.
nal ear canal; a long-term tympanostomy tube allows MD has a high impact on health-related quality of
transmission of the pressure pulses from the ear canal life. The unexpected spells of vertigo and the communi-
to the inner-ear fluid system via the round- and oval- cation problems related to poor speech discrimination
window membranes. The therapy consists of a 6-week are major factors that restrict daily routines (Yardley
trial period with three daily sessions for 5 minutes. Otor- et al., 2003).
rhea associated with the tympanostomy tube is the most Anxiety and depression are common comorbidities in
common adverse effect. Two recent meta-analyses show patients with MD (S€oderman et al., 2002; van Cruijsen
conflicting results (Ahsan et al., 2015; Syed et al., 2015). et al., 2006). The mental health impact of MD is
inversely related to time once it has been diagnosed
Hearing aids (Tyrrell et al., 2014). The majority of patients with
MD report psychologic stress as one of the main triggers
When hearing disability begins to affect quality of life, of their attacks (Kirby and Yardley, 2012). Validated
hearing aids should be advised. Fitting amplification to questionnaires can be used to detect behavioral factors
patients with MD involves several problems: fluctuating in vestibular patients (Staab, 2013). Relaxation tech-
nature of hearing loss, low-frequency hearing loss, uni- niques and cognitive behavioral therapy can help
lateral or asymmetric hearing loss, reduced dynamic patients to cope with their symptoms (Yardley and
range, and reduced word recognition scores (Valente Kirby, 2006). Self-help groups can also give patients
et al., 2006; McNeill et al., 2008). Modern digital hearing the opportunity to support each other (Porter and
aids can be specifically programmed to take these chal- Boothroyd, 2015).
lenges into account. Tinnitus is a common complaint in
advanced stages of MD when hearing loss no longer fluc-
tuates and becomes permanent. In these patients, hearing GUIDELINE FOR CONSERVATIVE
aids provide not only better speech discrimination, but TREATMENT OF MD
also sound enrichment that may facilitate the process
of habituation to their tinnitus. Lifestyle modifications and medical therapy are able to
control vestibular symptoms in most patients, although
VESTIBULAR REHABILITATION they have no effect on progression of hearing loss or tin-
nitus. We have developed a stepwise approach based on
Commonly, the role of vestibular rehabilitation in MD the number of episodes of vertigo and hearing level
has been limited in improving stability in patients with which can effectively control vertigo in most patients
nonfluctuating complete unilateral vestibular loss after a without surgery (Fig. 19.2). Conservative treatment
vestibular neurectomy or labyrinthectomy, and in treat- includes the following steps:
ing unsteadiness after IGT. A second indication is in
individuals with unsteadiness and disequilibrium in 1. low-sodium diet (around 1800 mg/day) and high
which vertigo spells are controlled by oral medication water intake (2000 mL/day), considered as baseline
or IGT or have ceased at a later stage (Gottshall therapy
et al., 2005). 2. betahistine 24 mg/8 hours for at least 6 months
Aside from these indications, the use of vestibular 3. prednisone 1 mg/kg for 15 days if multiple episodes
rehabilitation in patients with fluctuating vestibular loss of vertigo with longer duration are observed in con-
is not well established (Gottshall et al., 2010). In addi- secutive weeks or a sudden drop in hearing level is
tion, it is unclear in patients with bilateral MD. found. If no response is observed, prednisone is
stopped in 4 weeks
4. intratympanic gentamicin, if there are six episodes in
EDUCATIONAL AND PSYCHOLOGIC
the last 3 months. The most studied dosage is
ASPECTS
0.3–0.5 mL gentamicin sulfate using a concentra-
It is necessary to enquire into the concerns of patients and tion of 26.4 mg/mL. It can be repeated up to four
identify their fears, uncertainties, beliefs, and expecta- times. Gentamicin should not be used in patients
tions. It is essential to inform patients about the natural with bilateral disease, since they can develop bilat-
history of MD, to clarify any aspect related to the disor- eral vestibular hypofunction and persistent vestibu-
der and its therapy (Kirby and Yardley, 2009; Arroll et al., lar ataxia.
270 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
Define N᎑⬚ episodes >20 min
during last 3 mo (Vo)
N᎑⬚ episodes <50% Vo

Start with low Na* diet and


low Na+ water > 2l/d x 3m Yes No

N᎑⬚ episodes <50% Vo Betahistine* 24 mg/12 h x 6m


*Stop after 1 year w/o episodes of vertigo
Yes
Prednisone 1mg/kg/d PO Resp += improve 15 dB PTA
No
x15d, tapering in 4 wk o N᎑⬚ episodes <50% Vo

Caloric test/
No response Grommets
VEMPs/ vHIT

IT Gentamicin
Normal Unilateral
At least 6 episodes in the last 3 mo
response vestibular 0.3-0.5 ml @26,4 mg/ml until:
hypofunction Spontaneous Nystagmus
HIT +
Reconsider No episodes If vertigo relapses
diagnosis Repeat ITG until 4 times

Fig. 19.2. Stepwise approach to conservative management of Menière’s disease based on the number of episodes of vertigo and
hearing thresholds. VEMPs, vestibular-evoked myogenic potentials; vHIT, video-head impulse test; IGT, intratympanic gentami-
cin therapy; PTA, pure-tone audiometry. (Modified after Lopez-Escamez and Espinosa-Sanchez, 2014.)

NEW PERSPECTIVES generated from peripheral blood mononuclear cells or


skin fibroblasts of patients with MD (Almeida-Branco
New techniques for intratympanic delivery of drugs for
et al., 2015).
inner-ear absorption have been developed (Lambert
et al., 2012). These include hydrogel application and
REFERENCES
nanoparticles of poly-lactic/glycocolic acid; nanoparti-
cles are useful when a sustained release is intended. To Adrion C, Fischer CS, Wagner J et al. (2016). Betahistine ther-
obviate the inherent problems associated with middle- apy in patients with Menière’s disease: primary results of a
ear application, newer procedures are been investigated long-term, multicenter, double-blind, randomized, placebo-
to deliver medication directly to the inner ear. Cochlear controlled, dose-defining trial of efficacy and safety
implants, which are placed into the scala tympani, could (BEMED trial). Br Med J 352: h6816.
Ahsan SF, Standring R, Wang Y (2015). Systematic review
be a method for drug delivery directly to the inner ear.
and meta-analysis of Meniett therapy for Menière’s dis-
In vitro and in vivo experiments in animals have achieved
ease. Laryngoscope 125: 203–208.
delivery of dexamethasone and neurotrophic factors Albu S, Chirtes F, Trombitas V et al. (2015). Intratympanic
(brain-derived neurotrophic factor, neurotrophin-3) by dexamethasone versus high dosage of betahistine in the
a modification of the electrode. Moreover, direct inner- treatment of intractable unilateral Menière disease. Am
ear delivery via an osmotic pump with a perfusion system J Otolaryngol 36: 205–209.
has been developed in animal models. This last approach Alles MJ, der Gaag MA, Stokroos RJ (2006). Intratympanic
is based on microfluidics and miniaturization technolo- steroid therapy for inner ear diseases, a review of the liter-
gies and has the advantage of greater control and preci- ature. Eur Arch Otorhinolaryngol 263: 791–797.
sion of the delivery process. Almeida-Branco MS, Cabrera S, Lopez-Escamez JA (2015).
These new direct inner-ear delivery systems also have Perspectives for the treatment of sensorineural hearing
loss by cellular regeneration of the inner ear. Acta
been investigated with the purpose of gene therapy, like
Otorrinolaringol Esp 66: 286–295.
RNA interference techniques to silence target genes, or
Al-Momani MO, Ferraro JA, Gajewski BJ et al. (2009).
gene edition to repair mutations. Another field of animal Improved sensitivity of electrocochleography in the diag-
experimentation is stem cell therapy for hearing restora- nosis of Menière’s disease. Int J Audiol 48: 811–819.
tion, either promoting transdifferentiation of supporting Andrews JC, Honrubia V (2010). Premenstrual exacerbation
cells of the organ of Corti into hair cells or using gene- of Menière’s disease revisited. Otolaryngol Clin North
edited induced pluripotent stem cells, which could be Am 43: 1029–1240.
MENIÈRE’S DISEASE 271
Arroll M, Dancey CP, Attree EA et al. (2012). People with Cawthorne T, Hewlett AB (1954). Menière’s disease. Proc
symptoms of Menière’s disease: the relationship between R Soc Med 47 (8): 663–670.
illness intrusiveness, illness uncertainty, dizziness handi- Celestino D, Ralli G (1991). Incidence of Menière’s disease in
cap, and depression. Otol Neurotol 33: 816–823. Italy. Am J Otol 12: 135–138.
Arweiler-Harbeck D, Horsthemke B, Jahnke K et al. (2011). Cha YH, Brodsky J, Ishiyama G et al. (2007). The relevance of
Genetic aspects of familial Menière’s disease. Otol migraine in patients with Me nière’s disease. Acta
Neurotol 32 (4): 695–700. Otolaryngol 127: 1241–1245.
Ballester M, Liard P, Vibert D et al. (2002). Menière’s disease Charpiot A, Rohmer D, Gentine A (2010). Lateral semicircular
in the elderly. Otol Neurotol 23: 73–78. canal plugging in severe Menière’s disease: a clinical pro-
Banks C, McGinness S, Harvey R et al. (2012). Is allergy spective study about 28 patients. Otol Neurotol 31:
related to Menière’s disease. Curr Allergy Asthma Rep 237–240.
12: 255–260. Chávez H, Vega R, Soto E (2005). Histamine (H3) receptors
Belinchon A, Perez-Garrigues H, Tenias JM et al. (2011). modulate the excitatory amino acid receptor response of
Hearing assessment in Menière’s disease. Laryngoscope the vestibular afferents. Brain Res 1064: 1–9.
121: 622–626. Choung YH, Taura A, Pak K et al. (2009). Generation of
Belinchon A, Perez-Garrigues H, Tenias JM (2012). Evolution highly-reactive oxygen species is closely related to hair cell
of symptoms in Menière’s disease. Audiol Neurootol 17: damage in rat organ of Corti treated with gentamicin.
126–132. Neuroscience 161 (1): 214–226.
Benecke H, Perez-Garrigues H, Bin Sidek D et al. (2010). Claes GM, De Valck CF, Van de Heyning P et al. (2011). The
Effects of betahistine on patient-reported outcomes in rou- Menière’s disease index: an objective correlate of
tine practice in patients with vestibular vertigo and Menière’s disease based on audiometric and electroco-
appraisal of tolerability: experience in the OSVaLD study. chleographic data. Otol Neurotol 32: 887–892.
Int Tinnitus J 16: 14–24. Cohen-Kerem R, Kisilevsky V, Einarson TR et al. (2004).
Bergquist F, Ruthven A, Ludwig M et al. (2006). Intratympanic gentamicin for Menière’s disease: a meta-
Histaminergic and glycinergic modulation of GABA analysis. Laryngoscope 114: 2085–2091.
release in the vestibular nuclei of normal and labyrinthec- Colletti V, Carner M, Colletti L (2007). Auditory results after
tomised rats. J Physiol 577 (Pt 3): 857–868. vestibular nerve section and intratympanic gentamicin for
Bertlich M, Ihler F, Sharaf K et al. (2014). Betahistine metab- Menière’s disease. Otol Neurotol 28: 145–151.
olites, aminoethylpyridine, and hydroxyethylpyridine Committee on Hearing and Equilibrium guidelines for the
increase cochlear blood flow in guinea pigs in vivo. Int diagnosis and evaluation of therapy in Menière’s disease.
J Audiol 53: 753–759. American Academy of Otolaryngology-Head and Neck
odow A, Heinze M, Bloching MB et al. (2014). Caloric stim-
Bl€ Foundation, Inc (1995). Otolaryngol Head Neck Surg
ulation and video-head impulse testing in Menière’s dis- 113: 181–185.
ease and vestibular migraine. Acta Otolaryngol 134: Derebery JM (2000). Allergic management of Menière’s dis-
1239–1244. ease: an outcome study. Otolaryngol Head Neck Surg 122:
Boleas-Aguirre MS, Lin FR, Della Santina CC et al. (2008). 174–182.
Longitudinal results with intratympanic dexamethasone Derebery MJ (2011). Allergic and immunologic features of
in the treatment of Menière’s disease. Otol Neurotol 29: Menière’s disease. Otolaryngol Clin North Am 44:
33–38. 655–666.
Botta L, Mira E, Valli S et al. (1998). Effects of betahistine on Derebery MJ, Berliner KI (2010). Allergy and its relation to
vestibular receptors of the frog. Acta Otolaryngol 118: Menière’s disease. Otolaryngol Clin North Am 43:
519–523. 1047–1058.
Brantberg K, Baloh RW (2011). Similarity of vertigo attacks Desmadryl G, Gaboyard-Niay S, Brugeaud A et al. (2012).
due to Menière’s disease and benign recurrent vertigo, both Histamine H4 receptor antagonists as potent modulators
with and without migraine. Acta Otolaryngol 131: of mammalian vestibular primary neuron excitability. Br
722–727. J Pharmacol 167: 905–916.
Cabrera S, Sanchez E, Requena T et al. (2014). Intronic vari- Dobie RA, Snyder JM, Donaldson JA (1982).
ants in the NFKB1 gene may influence hearing forecast in Electronystagmographic and audiologic findings in patients
patients with unilateral sensorineural hearing loss in with Menière’s disease. Acta Otolaryngol 94: 19–27.
Menière’s disease. PLoS One 9. e112171. uwel P, J€
D€ ungling E, Westhofen M et al. (2003). Potassium
Casani AP, Piaggi P, Cerchiai N et al. (2012). Intratympanic currents in vestibular type II hair cells activated by hydro-
treatment of intractable unilateral Menière disease: genta- static pressure. Neuroscience 116: 963–972.
micin or dexamethasone? A randomized controlled trial. Dziadziola JK, Laurikainen EL, Rachel JD et al. (1999).
Otolaryngol Head Neck Surg 146: 430–437. Betahistine increases vestibular blood flow. Otolaryngol
Casani AP, Cerchiai N, Navari E et al. (2014). Intratympanic Head Neck Surg 120: 400–405.
gentamicin for Menière’s disease: short- and long-term Egami N, Kakigi A, Sakamoto T et al. (2013). Morphological
follow-up of two regimens of treatment. Otolaryngol and functional changes in a new animal model of Menière’s
Head Neck Surg 150: 847–852. disease. Lab Invest 93: 1001–1111.
272 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
Eklund S (1999). Headache in Menière’s disease. Auris Nasus Gottshall KR, Topp SG, Hoffer ME (2010). Early vestibular
Larynx 26: 427–433. physical therapy rehabilitation for Menière’s disease.
Espinosa-Sanchez JM, Heitzmann-Hernández T, López- Otolaryngol Clin North Am 43: 1113–1119.
Escámez JA (2014a). Tratamiento farmacológico de los Greco A, Gallo A, Fusconi M et al. (2012). Menière’s disease
acúfenos: mucho ruido y pocas nueces. Rev Neurol 59: might be an autoimmune condition? Autoimmun Rev 11:
164–174. 731–738.
Espinosa-Sanchez JM, Martin-Sierra C, Lopez-Escamez JA Green Jr JD, Blum DJ, Harner SG (1991). Longitudinal fol-
(2014b). Menière’s syndrome and migraine. In: lowup of patients with Menière’s disease. Otolaryngol
B Colombo, R Teggi (Eds.), Vestibular Migraine and Head Neck Surg 104: 783–788.
Related Syndromes, Springer International Publishing, Gu X, Fang ZM, Liu Y et al. (2015). Diagnostic advantages of
Heidelberg, pp. 129–141. intratympanically gadolinium contrast-enhanced magnetic
Ferraro JA, Durrant JD (2006). Electrocochleography in the resonance imaging in patients with bilateral Menière’s
evaluation of patients with Menière’s disease/endolym- disease. Am J Otolaryngol 36: 67–73.
phatic hydrops. J Am Acad Audiol 17: 45–68. Hamid M, Trune D (2008). Issues, indications, and controver-
Fife TA, Lewis MP, May JS et al. (2014). Cochlear implanta- sies regarding intratympanic steroid perfusion. Curr Opin
tion in Menière’s disease. JAMA Otolaryngol Head Neck Otolaryngol Head Neck Surg 16: 434–440.
Surg 140: 535–539. Hansen MR, Gantz BJ, Dunn C (2013). Outcomes after
Fiorino F, Pizzini FB, Beltramello A et al. (2011). Reliability cochlear implantation for patients with single-sided deaf-
of magnetic resonance imaging performed after intratym- ness, including those with recalcitrant Menière’s disease.
panic administration of gadolinium in the identification Otol Neurotol 34: 1681–1687.
of endolymphatic hydrops in patients with Menière’s dis- Harcourt J, Barraclough K, Bronstein AM (2014). Menière’s
ease. Otol Neurotol 32: 472–477. disease. BMJ 349: g6544.
Fisher LM, Derebery MJ, Friedman RA (2012). Oral steroid Harris JP, Alexander TH (2010). Current-day prevalence of
treatment for hearing improvement in Menière’s disease Menière’s syndrome. Audiol Neurootol 15: 318–322.
and endolymphatic hydrops. Otol Neurotol 33: 1685–1691. Havia M, Kentala E, Pyykk€ o I (2005). Prevalence of Menière’s
Fukushima M, Kitahara T, Fuse Y et al. (2004). Changes in disease in general population of Southern Finland.
aquaporin expression in the inner ear of the rat after i.p. Otolaryngol Head Neck Surg 133: 762–768.
injection of steroids. Acta Otolaryngol Suppl 553: 13–18. Herraiz C, Plaza G, Aparicio JM et al. (2010). Transtympanic
Gabra N, Saliba I (2013). The effect of intratympanic methyl- steroids for Menière’s disease. Otol Neurotol 31: 162–167.
prednisolone and gentamicin injection on Menière’s dis- Hietikko E, Kotim€aki J, Okuloff A et al. (2012). A replication
ease. Otolaryngol Head Neck Surg 148: 642–647. study on proposed candidate genes in Menière’s disease,
Gabriková D, Frykholm C, Friberg U et al. (2010). Familiar and a review of the current status of genetic studies. Int
Menière’s disease restricted to 1.48 Mb on chromosome J Audiol 51: 841–845.
12p12.3 by allelic and haplotype association. J Hum Hietikko E, Kotim€aki J, Sorri M et al. (2013). High incidence
Genet 55: 834–837. of Menière-like symptoms in relatives of Menière patients
Garduño-Anaya MA, Couthino De Toledo H, Hinojosa- in the areas of Oulu University Hospital and Kainuu Central
Gonzalez R et al. (2005). Dexamethasone inner ear perfu- Hospital in Finland. Eur J Med Genet 56: 279–285.
sion by intratympanic injection in unilateral Menière’s Hillman TA, Chen DA, Arriaga MA (2004). Vestibular nerve
disease: a two-year prospective, placebo-controlled, section versus intratympanic gentamicin for Menière’s
double-blind, randomized trial. Otolaryngol Head Neck disease. Laryngoscope 114: 216–222.
Surg 133: 285–294. House JW, Doherty JK, Fisher LM et al. (2006). Menière’s
Gazquez I, Lopez-Escamez JA (2011). Genetics of recurrent ver- disease: prevalence of contralateral ear involvement.
tigo and vestibular disorders. Curr Genomics 12: 443–450. Otol Neurotol 27: 355–361.
Gazquez I, Soto-Varela A, Aran I et al. (2011). High preva- Hu A, Parnes LS (2009). Intratympanic steroids for inner ear
lence of systemic autoimmune diseases in patients with disorders: a review. Audiol Neurootol 14: 373–382.
Menière’s disease. PLoS One 6. e26759. Hu A, Parnes LS (2010). 10-year review of endolymphatic sac
Gazquez I, Moreno A, Aran I et al. (2012). MICA-STR A.4 is surgery for intractable Menière disease. J Otolaryngol
associated with slower hearing loss progression in patients Head Neck Surg 39: 415–421.
with Menière’s disease. Otol Neurotol 33: 223–229. Huang CH, Wang SJ, Young YH (2011). Localization and
Gbahou F, Davenas E, Morisset S et al. (2010). Effects of beta- prevalence of hydrops formation in Menière’s disease
histine at histamine H3 receptors: mixed inverse agonism/ using a test battery. Audiol Neurootol 16: 41–48.
agonism in vitro and partial inverse agonism in vivo. Hulshof JH, Baarsma EA (1981). Vestibular investigations in
J Pharmacol Exp Ther 334: 945–954. Menière’s disease. Acta Otolaryngol 92: 75–81.
Ge X, Shea Jr JJ (2002). Transtympanic electrocochleography: Huon LK, Fang TY, Wang PC (2012). Outcomes of intratym-
a 10-year experience. Otol Neurotol 23: 799–805. panic gentamicin injection to treat Menière’s disease. Otol
Gottshall KR, Hoffer ME, Moore RJ et al. (2005). The role Neurotol 33: 706–714.
of vestibular rehabilitation in the treatment of Menière’s Huppert D, Strupp M, Brandt T (2010). Long-term course of
disease. Otolaryngol Head Neck Surg 133: 326–328. Menière’s disease revisited. Acta Otolaryngol 130: 644–651.
MENIÈRE’S DISEASE 273
Ihler F, Bertlich M, Sharaf K et al. (2012). Betahistine exerts a given as a single intratympanic injection in patients with
dose-dependent effect on cochlear stria vascularis blood unilateral Menière’s disease. Otol Neurotol 33: 1257–1265.
flow in guinea pigs in vivo. PLoS One 7. e39086. Lamounier P, Gobbo DA, de Souza TS et al. (2014).
James AL, Burton MJ (2001). Betahistine for Menière’s Electrocochleography for Menière’s disease: is it reliable?
disease or syndrome. Cochrane Database Syst Rev 1. Braz J Otorhinolaryngol 80: 527–532.
CD001873. Laurikainen E, Miller JF, Pyykk€ o I (2000). Betahistine effects
Jeck-Thole S, Wagner W (2006). Betahistine: a retrospective on cochlear blood flow: from the laboratory to the clinic.
synopsis of safety data. Drug Saf 29: 1049–1059. Acta Otolaryngol Suppl 544: 5–7.
Jerin C, Berman A, Krause E et al. (2014). Ocular vestibular Lee CS, Paparella MM, Margolis RH et al. (1995).
evoked myogenic potential frequency tuning in certain Audiological profiles and Menière’s disease. Ear Nose
Menière’s disease. Hear Res 310: 54–59. Throat J 74: 527–532.
Katayama N, Yamamoto M, Teranishi M et al. (2010). Lempert T, Olesen J, Furman J et al. (2012). Vestibular
Relationship between endolymphatic hydrops and migraine: diagnostic criteria. J Vestib Res 22: 167–172.
vestibular-evoked myogenic potential. Acta Otolaryngol Levo H, Kentala E, Rasku J et al. (2014). Aural fullness in
130: 917–923. Menière’s disease. Audiol Neurootol 19: 395–399.
Keles E, Godekmerdan A, Kalidag T et al. (2004). Menière’s Lezius F, Adrion C, Mansmann U et al. (2011). High-dosage
disease and allergy: allergens and cytokines. J Laryngol betahistine dihydrochloride between 288 and 480 mg/day
Otol 118: 688–693. in patients with severe Menière’s disease: a case series.
Kentala E, Havia M, Pyykk€o I (2001). Short-lasting drop Eur Arch Otorhinolaryngol 268: 1237–1240.
attacks in Menière’s disease. Otolaryngol Head Neck Li H, Feng G, Wang H, Feng Y (2015). Intratympanic steroid
Surg 124: 526–530. therapy as a salvage treatment for sudden sensorineural
Kim HH, Kumar A, Battista RA et al. (2005). hearing loss after failure of conventional therapy: a meta-
Electrocochleography in patients with Menière’s disease. analysis of randomized, controlled trials. Clin Ther 37:
Am J Otolaryngol 26: 128–131. 178–187.
Kim SH, Kim JY, Lee HJ et al. (2014). Autoimmunity as a Lin JS, Sakai K, Vanni-Mercier G et al. (1990). Involvement of
candidate for the etiopathogenesis of Menière’s disease: histaminergic neurons in arousal mechanisms demon-
detection of autoimmune reactions and diagnostic bio- strated with H3-receptor ligands in the cat. Brain Res
marker candidate. PLoS One 9. e111039. 523: 325–330.
King EB, Salt AN, Kel GE et al. (2013). Gentamicin adminis- Lopez-Escamez JA, Espinosa-Sanchez JM (2014). Opciones
tration on the stapes footplate causes greater hearing loss terapeuticas en la enfermedad de Menière. Actual Med
and vestibulotoxicity than round window administration 99 (Suppl 791): 27–32.
in guinea pigs. Hear Res 304: 159–166. Lopez-Escamez JA, López-Nevot A, Cortes R et al. (2002).
Kirby SE, Yardley L (2009). Cognitions associated with anx- Expression of A, B, C and DR antigens in definite
iety in Menière’s disease. J Psychosom Res 66: 111–118. Menière’s disease in a Spanish population. Eur Arch
Kirby SE, Yardley L (2012). Physical and psychological trig- Otorhinolaryngol 259: 347–350.
gers for attacks in Menière’s disease: the patient perspec- Lopez-Escamez JA, Vilchez JR, Soto-Varela A et al. (2007).
tive. Psychother Psychosom 81: 396–398. HLA-DRB1*1101 allele may be associated with bilateral
Kitahara T, Horii A, Imai T et al. (2014). Does endolymphatic Meniere’s disease in southern European population. Otol
sac decompression surgery prevent bilateral development Neurotol 28: 891–895.
of unilateral Menière disease? Laryngoscope 124: Lopez-Escamez JA, Viciana D, Garrido-Fernandez P (2009).
1932–1936. Impact of bilaterality and headache in health-related qual-
Klar J, Frykholm C, Friberg U et al. (2006). A Menière’s dis- ity of life in Menière’s disease. Ann Otol Rhinol Laryngol
ease gene linked to chromosome 12p12.3. Am J Med Genet 118: 409–416.
B Neuropsychiatr Genet 141B: 463–467. Lopez-Escamez JA, Saenz-Lopez P, Acosta L et al. (2010).
Kotim€aki J, Sorri M, Aantaa E et al. (1999). Prevalence Association of a functional polymorphism of PTPN22
of Menière disease in Finland. Laryngoscope 109: encoding a lymphoid protein phosphatase in bilateral
748–753. Menière’s disease. Laryngoscope 120: 103–107.
Krombach GA, van den Boom M, Di Martino E et al. (2005). Lopez-Escamez JA, Dlugaiczyk J, Jacobs J et al. (2014).
Computed tomography of the inner ear: size of anatomical Accompanying symptoms overlap during attacks in
structures in the normal temporal bone and in the temporal Menière’s disease and vestibular migraine. Front Neurol
bone of patients with Menière’s disease. Eur Radiol 15: 5: 265.
1505–1513. Lopez-Escamez JA, Carey J, Chung WH et al. (2015).
Lacour M (2013). Betahistine treatment in managing vertigo Diagnostic criteria for Menière’s disease. J Vestib Res
and improving vestibular compensation: clarification. 25: 1–7.
J Vestib Res 23: 139–151. Luxford E, Berliner KI, Lee J et al. (2013). Dietary modifica-
Lambert PR, Nguyen S, Maxwell KS et al. (2012). tion as adjunct treatment in Menière’s disease: patient
A randomized, double-blind, placebo-controlled clinical willingness and ability to comply. Otol Neurotol 34:
study to assess safety and clinical activity of OTO-104 1438–1443.
274 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
Lyford-Pike S, Vogelheim C, Chu E et al. (2007). Gentamicin Miyashita T, Toyama Y, Inamoto R et al. (2012). Evaluation of
isprimarily localized in vestibular type I hair cells after the vestibular aqueduct in Menière’s disease using multi-
intratympanic administration. J Assoc Res Otolaryngol 8: planar reconstruction images of CT. Auris Nasus Larynx
497–508. 39: 567–571.
MacDougall HG, Weber KP, McGarvie LA et al. (2009). The Moon IJ, Park GY, Choi J et al. (2012). Predictive value of
video head impulse test: diagnostic accuracy in peripheral electrocochleography for determining hearing outcomes
vestibulopathy. Neurology 73: 1134–1141. in Menière’s disease. Otol Neurotol 33: 204–210.
MacKeith SA, Bottrill LD, Ramsden JD (2014). Simultaneous Morales-Angulo C, Gallo-Terán J (2005). Crisis otolı́ticas de
labyrinthectomy with cochlear implantation in patients Tumarkin o drop attacks en pacientes con enfermedad de
with bilateral Menière’s disease. Ann Otol Rhinol Menière. Acta Otorrinolaringol Esp 56: 469–471.
Laryngol 123: 485–489. Morales-Luckie E, Cornejo-Suarez A, Zaragoza-Contreras
Manrique-Huarte R, Guillen-Grima F, Perez-Fernandez N MA et al. (2005 Sep). Oral administration of prednisone
(2011). Treatment of Menière’s disease with “on-demand” to control refractory vertigo in Menière’s disease: a pilot
intratympanic gentamicin injections. Otol Neurotol 32: study. Otol Neurotol 26 (5): 1022–1026.
461–465. Morrison AW, Bailey ME, Morrison GA (2009). Familial
Manzari L, Burgess AM, MacDougall HG et al. (2011). Rapid Menière’s disease: clinical and genetic aspects.
fluctuations in dynamic semicircular canal function in early J Laryngol Otol 123: 29–37.
Menière’s disease. Eur Arch Otorhinolaryngol 268: Murofushi T, Nakahara H, Yoshimura E et al. (2011).
637–639. Association of air-conducted sound oVEMP findings with
Marques PS, Perez-Fernandez N (2012). Bedside vestibular cVEMP and caloric test findings in patients with unilateral
examination in patients with unilateral definite Menière’s peripheral vestibular disorders. Acta Otolaryngol 131:
disease. Acta Otolaryngol 132: 498–504. 945–950.
Marques P, Manrique-Huarte R, Perez-Fernandez N (2015). Naganawa S, Komada T, Fukatsu H et al. (2006). Observation
Single intratympanic gentamicin injection in Menière’s of contrast enhancement in the cochlear fluid space of
disease: VOR change and prognostic usefulness. healthy subjects using a 3D-FLAIR sequence at 3 Tesla.
Laryngoscope 125: 1915–1920. Eur Radiol 16: 733–737.
Martens C, Csillag A, Davies M et al. (2013). Cochlear implan- Naganawa S, Satake H, Kawamura M et al. (2008). Separate
tation after selective vestibular nerve section. J Laryngol visualization of endolymphatic space, perilymphatic space
Otol 127: 311–313. and bone by a single pulse sequence; 3D-inversion recov-
Martin-Sanz E, Luzardo CZ, Riesco LR et al. (2013a). The use ery imaging utilizing real reconstruction after intratympa-
of electrocochleography to monitor the response of nic Gd-DTPA administration at 3 Tesla. Eur Radiol 18:
Menière’s disease patients to intratympanic steroids. 920–924.
Acta Otolaryngol 133: 1158–1164. Naganuma H, Kawahara K, Tokumasu K et al. (2006). Water
Martin-Sanz E, Zschaeck C, Gonzalez M et al. (2013b). may cure patients with Menière disease. Laryngoscope
Control of vertigo after intratympanic corticoid therapy 116: 1455–1460.
for unilateral Menière’s disease: a comparison of weekly Nakashima T, Naganawa S, Sugiura M et al. (2007).
versus daily fixed protocols. Otol Neurotol 34 (8): Visualization of endolymphatic hydrops in patients with
1429–1433. Menière’s disease. Laryngoscope 117: 415–420.
McCall AA, Swan EE, Borenstein JT et al. (2010). Drug deliv- Nauta JJ (2014). Meta-analysis of clinical studies with betahis-
ery for treatment of inner ear disease: current state of tine in Menière’s disease and vestibular vertigo. Eur Arch
knowledge. Ear Hear 31: 156–165. Otorhinolaryngol 271: 887–897.
McNeill C, McMahon CM, Newall P et al. (2008). Hearing Neff BA, Staab JP, Eggers SD et al. (2012). Auditory and ves-
aids for Menière’s syndrome: implications of hearing fluc- tibular symptoms and chronic subjective dizziness in
tuation. J Am Acad Audiol 19: 430–434. patients with Menière’s disease, vestibular migraine, and
McRackan TR, Best J, Pearce EC et al. (2014a). Intratympanic Menière’s disease with concomitant vestibular migraine.
dexamethasone as a symptomatic treatment for Menière’s Otol Neurotol 33: 1235–1244.
disease. Otol Neurotol 35: 1638–1640. Nevoux J, Viengchareun S, Lema I et al. (2015).
McRackan TR, Gifford RH, Kahue CN et al. (2014b). Glucocorticoids stimulate endolymphatic water reabsorp-
Cochlear implantation in Menière’s disease patients. Otol tion in inner ear through aquaporin 3 regulation. Pflugers
Neurotol 35: 421–425. Arch 467: 1931–1943.
Merchant SN, Adams JC, Nadol JB (2005). Pathophysiology Ogawa Y, Otsuka K, Hagiwara A et al. (2015). Clinical study
of Menière’s syndrome: are symptoms caused by endolym- of tympanostomy tube placement for patients with intrac-
phatic hydrops? Otol Neurotol 26: 74–81. table Menière’s disease. J Laryngol Otol 129: 120–125.
Miller MW, Agrawal Y (2014). Intratympanic therapies Ohmen JD, White CH, Li X et al. (2013). Genetic evidence for
for Menière’s disease. Curr Otorhinolaryngol Rep 2: an ethnic diversity in the susceptibility to Menière’s dis-
137–143. ease. Otol Neurotol 34: 1336–1341.
Miller ME, Bykowski J (2014). Imaging analysis of Menière’s Okafor BC (1984). Incidence of Menière’s disease. J Laryngol
disease. Curr Otorhinolaryngol Rep 2: 152–161. Otol 98: 775–779.
MENIÈRE’S DISEASE 275
Otake H, Yamamoto H, Teranishi M et al. (2009). Cochlear Quaglieri S, Gatti O, Rebecchi E et al. (2014). Intratympanic
blood flow during occlusion and reperfusion of the anterior gentamicin treatment ‘as needed’ for Menière’s disease.
inferior cerebellar artery: effect of topical application of Long-term analysis using the Kaplan-Meier method. Eur
dexamethasone to the round window. Acta Otolaryngol Arch Otorhinolaryngol 271: 1443–1449.
129: 127–131. Radtke A, Lempert T, Gresty MA et al. (2002). Migraine and
Paparella MM, Mancini F (1985). Vestibular Menière’s dis- Menière’s disease: is there a link? Neurology 59: 1700–1704.
ease. Otolaryngol Head Neck Surg 93: 148–151. Radtke A, von Breverm M, Neuhauser H et al. (2012).
Paradis J, Hu A, Parnes LS (2013). Endolymphatic sac surgery Vestibular migraine: long-term follow-up of clinical symp-
versus intratympanic gentamicin for the treatment of toms and vestibulo-cochlear findings. Neurology 79:
intractable Menière’s disease: a retrospective review with 1607–1614.
survey. Otol Neurotol 34: 1434–1437. Rah YC, Han JJ, Park J et al. (2015). Management of intracta-
Park JJ, Chen YS, Westhofen M (2009). Menière’s disease and ble Menière’s disease after intratympanic injection of gen-
middle ear pressure: vestibular function after transtympa- tamicin. Laryngoscope 125: 972–978.
nic tube placement. Acta Otolaryngol 129: 1408–1413. Ramos-Macı́as A, Falcón-González JC, Manrique M et al.
Parker W (1995). Menière’s disease. Etiologic considerations. (2015). Cochlear implants as treatment option for unilateral
Arch Otolaryngol Head Neck Surg 121: 377–382. hearing loss, severe tinnitus and hyperacusis. Audiol
Parnes LS, Sun AH, Freeman DJ (1999). Corticosteroid phar- Neurotol (suppl 1): 60–66.
macokinetics in the inner ear fluids: an animal study fol- Requena T, Gazquez I, Moreno A et al. (2013). Allelic variants
lowed by clinical application. Laryngoscope 109: 1–17. in TLR10 gene may influence bilateral affectation and clin-
Pathak S, Goldofsky E, Vivas EX et al. (2011). IL-1b is over- ical course of Menière’s disease. Immunogenetics 65:
expressed and aberrantly regulated in corticosteroid nonre- 345–355.
sponders with autoimmune inner ear disease. J Immunol Requena T, Espinosa-Sanchez JM, Cabrera S et al. (2014).
186: 1870–1879. Familial clustering and genetic heterogeneity in
Perez-Fernandez N, Montes-Jovellar L, Cervera-Paz J et al. Menière’s disease. Clin Genet 85: 245–252.
(2010). Auditory and vestibular assessment of patients with Requena T, Cabrera S, Martı́n-Sierra C et al. (2015).
Menière’s disease who suffer Tumarkin attacks. Audiol Identification of two novel mutations in FAM136A and
Neurootol 15: 399–406. DTNA genes in autosomal-dominant familial Menière’s
Perez-Garrigues H, Lopez-Escamez JA, Perez P et al. (2008). disease. Hum Mol Genet 24: 1119–11126.
Time course of episodes of definitive vertigo in Menière’s Sajjadi H, Paparella MM (2008). Menière’s disease. Lancet
disease. Arch Otolaryngol Head Neck Surg 134: 372: 406–414.
1149–1154. Salt AN, Plontke SK (2009). Principles of local drug delivery
Perez-Garrigues H, Tulsidas-Mahtani B, Cavalle L et al. to the inner ear. Audiol Neurootol 14: 350–360.
(2015). A new approach to the treatment of the three symp- Samy RN, Houston L, Scott M et al. (2015). Cochlear implan-
toms of Menière’s disease: labyrinthectomy and cochlear tation in patients with Menière’s disease. Cochlear
implant in the same surgical procedure. Acta Implants Int 16: 208–212.
Otorrinolaringol Esp 66: e13–e14. Seo YJ, Kim J, Choi JY et al. (2013). Visualization of endo-
Phillips JS, Westerberg B (2011). Intratympanic steroids for lymphatic hydrops and correlation with audio-vestibular
Menière’s disease or syndrome. Cochrane Database Syst functional testing in patients with definite Menière’s dis-
Rev 7. CD008514. ease. Auris Nasus Larynx 40: 167–172.
Platt M, Dilwali S, Elackattu A et al. (2013). Mining immune Shin JE, Kim CH, Park HJ (2013). Vestibular abnormality in
epitopes in the inner ear. Otolaryngol Head Neck Surg 150: patients with Menière’s disease and migrainous vertigo.
460–463. Acta Otolaryngol 133: 154–158.
Porter M, Boothroyd RA (2015). Symptom severity, social Shojaku H, Watanabe Y, Fujisaka M et al. (2005).
supports, coping styles, and quality of life among individ- Epidemiologic characteristics of definite Menière’s disease
uals’ diagnosed with Menierè’s disease. Chronic Illn 11: in Japan. A long-term survey of Toyama and Niigata pre-
256–266. fectures. ORL J Otorhinolaryngol Relat Spec 67: 305–309.
Proctor LR (2000). Results of serial vestibular testing in uni- Silverstein H, Smouha E, Jones R (1989). Natural history vs
lateral Menière’s disease. Am J Otol 21: 552–558. surgery for Menière’s disease. Otolaryngol Head Neck
Pullens B, van Benthem PP (2011). Intratympanic gentamicin Surg 100: 6–16.
for Menière’s disease or syndrome. Cochrane Database Sismanis A (2010). Surgical management of common periph-
Syst Rev 16. CD008234. eral vestibular diseases. Curr Opin Otolaryngol Head Neck
Pullens B, Verschuur HP, van Benthem PP (2013). Surgery for Surg 18: 431–435.
Menière’s disease. Cochrane Database Syst Rev 2. Smith MR, Nelson AB, Du Lac S (2002). Regulation of firing
CD005395. response gain by calcium-dependent mechanisms in vestib-
Pyykk€o I, Nakashima T, Yoshida T et al. (2013). Menière’s ular nucleus neurons. J Neurophysiol 87: 2031–2042.
disease: a reappraisal supported by a variable latency of Smith WK, Sankar V, Pfleiderer AG (2005). A national survey
symptoms and the MRI visualization of endolymphatic amongst UK otolaryngologists regarding the treatment of
hydrops. BMJ Open 3 (2). Menière’s disease. J Laryngol Otol 119: 102–105.
276 J.M. ESPINOSA-SANCHEZ AND J.A. LOPEZ-ESCAMEZ
oderman AC, Bagger-Sj€ob€ack D, Bergenius J et al. (2002).
S€ Valente M, Mispagel K, Valente LM et al. (2006). Problems
Factors influencing quality of life in patients with and solutions for fitting amplification to patients with
Menière’s disease, identified by a multidimensional Menière’s disease. J Am Acad Audiol 17: 6–15.
approach. Otol Neurotol 23: 941–948. van Cruijsen N, Jaspers JP, van de Wiel HB et al. (2006).
Staab JP (2013). Behavioural neuro-otology. In: AM Bronstein Psychological assessment of patients with Menière’s dis-
(Ed.), Oxford textbook of vertigo and imbalance, Oxford ease. Int J Audiol 45: 496–502.
University Press, Oxford, pp. 333–346. Van Heyning P, Vermeire K, Diebl M et al. (2008).
Stahle J, Stahle C, Arenberg IK (1978). Incidence of Menière’s Incapacitating unilateral tinnitus in single-sided deafness
disease. Arch Otolaryngol 104: 99–102. treated by cochlear implantation. Ann Otol Rhinol
Stahle J, Friberg U, Svedberg A (1991). Long-term progres- Laryngol 117: 645–652.
sion of Meniere’s disease. Acta Otolaryngol Suppl 485: Vermeire K, Van de Heyning P (2009). Binaural Hearing
78–83. after cochlear implantation in subjects with unilateral
Strupp M, Hupert D, Frenzel C et al. (2008). Long-term sensorineural deafness and tinnitus. Audiol Neurotol 14:
prophylactic treatment of attacks of vertigo in Menière’s 163–171.
disease – comparison of a high with a low dosage of beta- Vermeire K, Van Yper L, De Vel E et al. (2014). Is cochlear
histine in an open trial. Acta Otolaryngol 128: 520–524. implantation an effective treatment for Menière’s disease?
Svrakic M, Pathak S, Goldofsky E et al. (2012). Diagnostic and B-ENT 10: 93–98.
prognostic utility of measuring tumor necrosis factor in the Viana LM, Bahmad Jr F, Rauch SD (2014). Intratympanic gen-
peripheral circulation of patients with immune-mediated tamicin as a treatment for drop attacks in patients with
sensorineural hearing loss. Arch Otolaryngol Head Neck Menière’s disease. Laryngoscope 124: 2151–2154.
Surg 138: 1052–1058. Vrabec JT (2010). Genetic investigations of Menière’s dis-
Syed MI, Rutka J, Hendry J et al. (2015). Positive pressure ease. Otolaryngol Clin North Am 43: 1121–1132.
therapy for Menière’s syndrome/disease with a Meniett Vrabec JT, Simon LM, Coker NJ (2007). Survey of Menière’s
device: a systematic review of randomised controlled trials. disease in a subspecialty referral practice. Otolaryngol
Clin Otolaryngol 40: 197–207. Head Neck Surg 137: 213–217.
Takeda T, Kakigi A (2010). The clinical value of extra- Wang HM, Tsai SM, Chien CY et al. (2012). Analysis
tympanic electrocochleography in the diagnosis of of auditory and vestibular function in patients with
Menière’s disease. ORL J Otorhinolaryngol Relat Spec unilateral Menière’s disease. Acta Otolaryngol 132:
72: 196–204. 1246–1251.
Tapia-Toca MC, Herraiz-Puchol C, Antonı́-Candela F (2009). Watanabe Y, Mizukoshi K, Shojaku H et al. (1995).
Tratamiento medico de la enfermedd de Menière. Epidemiological and clinical characteristics of Menière’s
In: JA Lopez-Escamez et al. (Eds.), Enfermedad de disease in Japan. Acta Otolaryngol Suppl 519: 206–210.
Menière: desde las ciencias básicas hacia la medicina Weinreich HM, Agrawal Y (2014). The link between allergy
clı́nica. Badalona, Euromedice, pp. 251–266. and Menière’s disease. Curr Opin Otolaryngol Head
Taylor RL, Wijewardene AA, Gibson WP et al. (2011). The Neck Surg 22: 227–230.
vestibular evoked potential profile of Menière’s disease. Wiet RJ (1979). Patterns of ear disease in the southwestern
Clin Neurophysiol 122: 1256–1263. American Indian. Arch Otolaryngol 105: 381–385.
Teufert KB, Doherty J (2010). Endolymphatic sac shunt, Winters SM, Campschroer T, Grolman et al. (2011). Ocular
labyrinthectomy, and vestibular nerve section in vestibular evoked myogenic potentials in response to air-
Menière’s disease. Otolaryngol Clin North Am 43: conducted sound in Menière’s disease. Otol Neurotol 32:
1091–1111. 1273–1280.
Thirlwall AS, Kundu S (2006). Diuretics for Menière’s disease Wladislavosky-Waserman P, Facer GW, Mokri B et al. (1984).
or syndrome. Cochrane Database Syst Rev 3. CD003599. Menière’s disease: a 30-year epidemiologic and clinical
Tighilet B, Trottier S, Lacour M (2005). Dose- and duration- study in Rochester, Mn, 1951–1980. Laryngoscope 94:
dependent effects of betahistine dihydrochloride treatment 1098–1102.
on histamine turnover in the cat. Eur J Pharmacol 523: Yardley L, Kirby S (2006). Evaluation of booklet-based self-
54–63. management of symptoms inMenière disease: a random-
Tighilet B, Mourre C, Trottier S et al. (2007). Histaminergic ized controlled trial. Psychosom Med 68: 762–769.
ligands improve vestibular compensation in the cat: beha- Yardley L, Dibb B, Osborne G (2003). Factors associated with
vioural, neurochemical and molecular evidence. Eur quality of life in Menière’s disease. Clin Otolaryngol Allied
J Pharmacol 568: 149–163. Sci 28: 436–441.
Tokumasu K, Fujino A, Naganuma H et al. (1996). Initial Yin S, Chen Z, Yu D et al. (2008). Triple semicircular canal
symptoms and retrospective evaluation of prognosis in occlusion for the treatment of Menière’s disease. Acta
Menière’s disease. Acta Otolaryngol Suppl 524: 43–49. Otolaryngol 128: 739–743.
Tyrrell JS, Whinney DJ, Ukoumunne OC et al. (2014). Young YH (2013). Potential application of ocular and cervical
Prevalence, associated factors, and comorbid conditions vestibular evoked myogenic potentials in Menière’s dis-
for Menière’s disease. Ear Hear 35: e162–e169. ease: a review. Laryngoscope 123: 484–491.
MENIÈRE’S DISEASE 277
Young YH, Wu CC, Wu CH (2002). Augmentation of vestib- Zhao R, Zhou H, Su SB (2013). A critical role for interleukin-
ular evoked myogenic potentials: an indication for 1b in the progression of autoimmune diseases. Int
distended saccular hydrops. Laryngoscope 112: 509–512. Immunopharmacol 17 (3): 658–669.
Young YH, Huang TW, Cheng PW (2003). Assessing the stage Zulueta-Santos C, Lujan B, Manrique-Huarte R et al. (2014).
of Menière’s disease using vestibular evoked myogenic The vestibulo-ocular reflex assessment in patients with
potentials. Arch Otolaryngol Head Neck Surg 129: Menière’s disease: examining all semicircular canals.
815–818. Acta Otolaryngol 134: 1128–1133.

You might also like