You are on page 1of 10

Rheumatology 2013;52:780–789

RHEUMATOLOGY doi:10.1093/rheumatology/ket009
Advance Access publication 21 February 2013

Review
Autoimmune sensorineural hearing loss: the
otology–rheumatology interface

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


Tamara Mijovic1, Anthony Zeitouni1 and Inés Colmegna2

Abstract
Autoimmune sensorineural hearing loss (SNHL) is a rare clinical entity characterized by a progressive
fluctuating bilateral asymmetric SNHL that develops over several weeks to months. Vestibular symptoms,
tinnitus and aural fullness are present in up to 50% of patients. Due to the lack of specific diagnostic tests,
both clinical suspicion and responsiveness to corticosteroids are the pillars for the diagnosis of autoim-
mune SNHL. The evaluation of patients in whom this condition is suspected should include a detailed
history and physical examination, an audiogram, an MRI and a limited laboratory workup to exclude
R EV I E W

secondary causes of hearing loss. The low frequency of this condition, the heterogeneity in the designs
of the available studies and the absence of randomized trials comparing treatment responses and
assessing long-term outcomes are some of the factors accounting for the limited evidence to guide the
clinician in the approach to the diagnosis and treatment of autoimmune SNHL.
Key words: hearing, deafness, autoimmune inner ear disease (AIED), sensorineural hearing loss (SNHL),
autoimmune cochleopathy, immune-mediated cochleovestibular disease, fluctuating hearing loss.

Introduction AIED accounts for fewer than 1% of all cases of hearing


loss; however, this could be an underestimation based
Sensorineural hearing loss (SNHL) results from dysfunc- on the absence of specific diagnostic tests [3]. AIED is
tion of the inner ear, the vestibulocochlear nerve or the one of the few forms of sensorineural deafness that can
central processing centres of the brain. In contrast, con- potentially be treated.
ductive hearing loss results from a defect in sound trans- Herein, we summarize the evidence on the pathophysi-
mission through the external ear, tympanic membrane or ology and current approach to the diagnosis and treat-
middle ear. ment of adults with bilateral, progressive, fluctuating
In 1958, Lehnhardt first described an ‘antigen–antibody hearing loss. These data derive from a systematic
reaction’ in 13 patients with progressive bilateral SNHL. In review of the literature conducted in MEDLINE (1996 to
his report, nine patients developed subsequent involve- October 2012) and EMBASE (1996 to October 2012).
ment of the contralateral ear, suggesting the possibility The search strategy included the terms autoimmune
that an injury on one side sensitized the body to a coch- and hearing loss both as medical subject heading and
lear antigen leading to hearing loss in the opposite ear [1]. keywords.
In 1979, McCabe [2] describes one patient from a series of
18 with idiopathic bilateral progressive SNHL responsive Definitions
to steroids, proposing the term autoimmune SNHL. This
condition is also known as autoimmune inner ear disease In the absence of a reliable marker for AIED, the disease
(AIED), autoimmune cochleopathy or immune-mediated is defined by an appropriate clinical presentation, by ex-
cochleovestibular disease [3, 4]. clusion of other known causes of SNHL and by a positive
response to steroid therapy.
1
McCabe’s original description states that the time
Department of Otolaryngology, Head and Neck Surgery and 2Section
of Rheumatology, Department of Medicine, McGill University, course of the hearing loss is the best clue to AIED diag-
Montreal, Quebec, Canada. nosis: ‘a period of progression of the deafness over weeks
Submitted 19 October 2012; revised version accepted or months, not hours nor days nor years’ [2]. This course
17 January 2013. distinguishes AIED from sudden deafness and from
Correspondence to: Inés Colmegna, Section of Rheumatology, age-related hearing loss (i.e. presbycusis). AIED is defined
Department of Medicine, Royal Victoria Hospital, M11-32, 687 Pine
Avenue, Montreal, Quebec H3A 1A1, Canada. as primary when the pathology is restricted to the ear. In
E-mail: ines.colmegna@mcgill.ca up to a third of cases, AIED occurs in the context of a

! The Author 2013. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For Permissions, please email: journals.permissions@oup.com
Autoimmune inner ear disease

TABLE 1 Systemic autoimmune diseases associated with cell-mediated mechanisms are involved in the autoim-
AIED mune injury to the inner ear. Cochlear innate immunity
has been proposed to contribute to the initiation of a
Disease
local adaptive immune response following antigen chal-
lenge. Briefly, as a consequence of a yet undefined trigger
Vogt–Koyanagi–Harada syndrome (i.e. antibody cross-reactivity, viral injury, trauma, vascular
Cogan’s syndrome insult, surgical damage and others) damaging the inner
Susac’s syndrome ear, lymphocytes from the systemic circulation are
SLE

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


exposed to proteins of the cochlea. Animals immunized
Antiphospholipid syndrome with inner ear homogenate (reviewed in [12]) develop an
RA
immune reaction in the endolymphatic sac, which is a
Systemic vasculitis
critical organ in processing and modulating immunity in
Panarteritis nodosa
Granulomatosis with polyangiitis the inner ear [13, 14]. Immune cells from the systemic
SSc circulation penetrate the blood–labyrinthine barrier to
Goodpasture syndrome reach the endolymphatic sac [3]. Specifically, the blood
Behçet’s disease vessels of the spiral modiolar vein adjacent to the scala
Sarcoidosis tympani seem to be the initial site for lymphocyte entry to
SS the inner ear [15]. Chronic activation of helper T lympho-
Hashimoto thyroiditis cytes reactive against self-proteins leads to the destruc-
Relapsing polychondritis tion of sensory and supporting cells within the cochlea.
Myasthenia gravis
IL-1a, IL-2, TNF-a and NF-kB, cytokines essential in the
Polymyositis–dermatomyositis
initiation, modulation and amplification of the immune
Crohn’s disease
Ulcerative colitis response, are expressed by infiltrating cells in the endo-
AS lymphatic sac [16, 17]. The pathogenic role of autoreac-
Guillain–Barré syndrome tive T-cell lines has been demonstrated by their passive
Multiple sclerosis transfer to naı̈ve animals resulting in an inner ear autoim-
mune response [18, 19]. Thus, a critical task is to identify
Adapted from Acta Otolaryngol 2006;126:1012–21. the inner ear antigens that trigger this autoreactive re-
sponse. A proposed candidate is recombinant mouse
systemic autoimmune disease and is defined as second- cochlin, an extracellular matrix protein highly and specif-
ary [5]. Table 1 provides a list of the systemic autoimmune ically expressed in the inner ear [20]. When challenged
diseases associated with AIED; the true frequency and the with cochlin antigen, mice have robust T-cell proliferative
risk factors for developing AIED in the context of systemic responses, increased production of IFN-g and cochlear
autoimmune diseases are unknown. inflammation [21]. Moreover, this form of primary AIED
could be passively transferred into naı̈ve recipient mice
Although there are no uniformly accepted criteria to
by CD4+ T cells. Of relevance, elevated levels of cochlin
diagnose AIED, the presence of bilateral SNHL of 30 dB
antibodies and increased frequencies of cochlin-specific
or more at any frequency with evidence of progression in
IFN-g-producing T cells have been found in patients with
at least one ear on two serial audiograms performed less
AIED [22, 23]. These studies provide evidence implicating
than 3 months apart is often used for case definition [6, 7].
an adaptive immune response, against an inner-ear-spe-
cific target antigen, with AIED. They also provide experi-
Pathophysiology mental confirmation that AIED may be a T-cell-mediated
organ-specific autoimmune disorder of the inner ear [21].
The pathogenesis of bilateral, progressive, fluctuating Further data supporting the role of T cells in AIED derives
hearing loss is unknown and therefore this represents from a murine study treated with adipose-derived mesen-
one of the most controversial and challenging diseases chymal stromal cells. This cell-based therapeutic strategy
in otology [8]. Since diagnostic biopsy of the human significantly improved hearing function via the down regu-
inner ear is not feasible, animal models are of major rele- lation of T-cell responses and the suppression of Th1/
vance in studying the aetiopathogenesis of this disease. Th17-type cytokines through the generation of IL-10-
Viral infections or vascular disorders leading to cochlear secreting T-reg cells [24].
or cochlear nerve damage have been postulated as Circulating antibodies against a number of inner ear anti-
mechanisms in bilateral, progressive, fluctuating hearing gens leading to cytotoxic antibody-mediated damage
loss. However, up to date, there is very limited evidence (type II response) or immune complex-mediated damage
to support the pathogenetic role of viral or vascular (type III response) have been implicated in the pathogen-
injuries [8]. esis of hearing loss in animal models. Sera from 70% of
The strongest pathogenic evidence is that of an patients with bilateral SSHL of unknown aetiology contain
immune-mediated disease. The inner ear is not an im- antibodies capable of immunostaining human inner ear
munologically privileged site and may mount an immune tissues. Autoantibodies against type II collagen, type IX
response against foreign and self-antigens damaging collagen, Raf-1 protein, the major peripheral myelin
sensory structures within it [9–11]. Both humoral and protein P0, B-actin, cochlin, choline transporter-like

www.rheumatology.oxfordjournals.org 781
Tamara Mijovic et al.

protein 2 (CTL2), cell-density enhanced protein tyrosine symptoms in the contralateral ear should undergo workup
phosphatase-1 and connexin 26 have all been described for an autoimmune aetiology [37]. As stated earlier, up to a
in AIED [4, 25, 26]. The stria vascularis, fibrocytes of the third of AIED cases occur in the context of a systemic auto-
spiral ligament and supporting cells are the anatomical immune disease, and unusually can be the first manifest-
targets of those autoantibodies. ation of the latter.
Harris and Sharp [27] were the first to show that western
blots of serum from patients with AIED had an antibody Diagnosis
directed against the 68-kDa inner ear antigen latter char-
Essentially all patients with AIED initially present to and are

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


acterized as a heat shock protein (HSP70) found in the
inner ear. Of relevance, elevated HSP70 antibodies are evaluated by otolaryngologists who often consult rheuma-
not the underlying cause of hearing loss but rather a non- tologists to rule out an underlying systemic autoimmune
pathogenetic epiphenomenon [28, 29]. However, antibo- disease or to assist in monitoring the immunosuppressive
dies to HSP70 are found in AIED patients and their therapy. Fig. 1 proposes a step-by-step diagnostic ap-
presence was correlated with responsiveness to steroid proach. The following features should be determined
treatment [6]. The sensitivity and specificity of anti-HSP70 from the history in any patient presenting with hearing
for the diagnosis of AIED was 42% and 90%, respectively, loss: (i) length of time over which the hearing loss has de-
and their positive predictive value 91% [30]. veloped, (ii) associated otological symptoms (tinnitus, ver-
In experimental models of AIED (i.e. the immunization of tigo, pain, discharge and pressure) and (iii) predisposing
a guinea pig with swine inner ear antigens in complete factors (i.e. noise exposure, chemotherapy or antibiotic
Freund’s adjuvant), it is possible to induce cochlear dys- treatments, previous ear surgery, trauma, meningitis or
function, endolymphatic hydrops and development of family history of hearing loss). Differentiating between
antibodies to 68-, 50-, 45- and 27-kDa molecular weight AIED and sudden hearing loss is critical. Although at the
inner antigens. The target antigen seems to be a choline initial presentation AIED may be more evident in one ear, it
transporter-like protein 2 (CTL2), a member of the cho- progresses over weeks to months, involving both ears. In
line transporter-like protein family linked to acetylcholine contrast, sudden hearing loss is unilateral and develops in
synthesis, which is an inner ear glycoprotein of 68 and 72 h or less. In addition to the specific otological history, it is
72 kDa [31]. relevant to perform a complete review of systems since in
In summary, both arms of the immune system, innate 15–30% of cases patients with AIED have or will develop a
and adaptive, are implicated in the processes ultimately systemic autoimmune disease (Table 1) [36]. Thus, the
leading to the histopathological features in the cochlea of presence of recurrent or chronic ocular disease, nephritis,
patients with AIED. Those include damage to the organ of arthritis, pneumonitis, sinusitis, photosensitivity and symp-
Corti, retrograde neural degeneration to the level of the toms suggestive of inflammatory bowel should be estab-
spiral ganglion, endolymphatic hydrops, stria vascularis lished. In addition to causing SNHL, autoimmune diseases
dystrophy, neo-fibroosteogenesis in the basal turn of the can lead to conductive hearing impairment. In fact, up to a
cochlea, fibrosis of the endolymphatic sac and lympho- third of patients with granulomatosis with polyangiitis have
cytes in the labyrinthine membrane compartment [32–35]. conductive hearing loss secondary to effusions from
granulomatous involvement of the middle ear and eusta-
Clinical manifestations chian tube mucosa [41]. Similarly, eustachian tube dys-
function is not uncommon in patients with relapsing
AIED most commonly affects people in the third to sixth polychondritis [42], while the ossicular chain can be af-
decades of life and presents with unilateral or bilateral fected by RA, leading to conductive hearing loss in more
SNHL that progresses over several weeks to months than 15% of RA patients [43].
[2, 36]. This time course of the hearing loss is the best Therefore, the first step in the evaluation of a patient
clue to the diagnosis. Although at presentation 50% of presenting with hypoacusia is to define whether the hear-
patients report unilateral hearing loss, audiological evalu- ing loss is sensorineural or conductive. This is based on
ation demonstrates asymmetrical but bilateral involvement the physical examination, specifically on the otoscopy
in the vast majority of cases (80–100%) [2]. Fluctuations in findings and tuning forks tests. The magnitude of hearing
hearing are common but the overall course is one of pro- loss is then defined by an audiogram where air and bone
gressive deterioration of auditory function [37–39]. conduction hearing thresholds are quantified and com-
Vestibular symptoms such as generalized imbalance, pared. Fig. 2 shows a representative example of an audio-
motion intolerance, ataxia and positional or episodic low- gram of conductive hearing loss and SNHL. The
grade vertigo are present in 50–80% of patients [3, 35, 37]; audiological assessment is a crucial part of both the initial
25–50% have tinnitus and aural fullness meeting criteria for diagnostic evaluation and all subsequent follow-ups of
Menière’s disease [37]. In fact, during the first months, the patients with hearing loss, allowing the characterization
differential diagnosis between Menière’s disease and AIED of the severity, frequencies involved and symmetry. The
can be difficult. The more aggressive course of AIED will two most important components of the audiological as-
allow the differentiation. Moreover, some forms of bilateral sessment in patients with AIED are (i) pure-tone air con-
Menière’s disease seem to have an underlying autoimmune duction threshold testing, the goal of which is to obtain a
pathology [40]. Some authors suggest that any patient representation of the softest intensity heard across fre-
diagnosed with unilateral Menière’s disease who develops quencies (0.25, 0.5, 1, 2, 3, 4, 6 and 8 kHz) comparing

782 www.rheumatology.oxfordjournals.org
Autoimmune inner ear disease

FIG. 1 Algorithm for the approach to a patient with hearing loss.

Duration (weeks to months)


Associated otological symptoms (tinnitus, vertigo, pain, discharge, pressure- 50% of patients)
History Rule out known causes of SNHL (noise exposure, medications, infections, trauma, surgery, family history)
Review of symptoms for autoimmune disorders (ocular, renal, pulmonary, ENT, GI and skin involvement)

Physical

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


Examination Otoscopy and tuning forks (SNHL)

Audiogram Asymmetrical SNHL

MRI Exclude retrocochlear pathology (no acoustic neuroma, multiple sclerosis, metastatic lesions or ischaemia)

Labs Rule out other causes of SNHL and associated autoimmune disorders (CBC, ESR, anti-dsDNA, anti-
SSA/B, antiphospholipid treponemal Ab absorption test)

Suspect
AIED

Consult OTL,
Response: continue steroids until
Trial of steroids Repeat audiogram stabilization and taper in 6 months
Rheumatology
& Audiology
(1 mg/kg/d x 4weeks)
No response: taper steroids over
10 days

OTL: otolaryngologist; CBC: complete blood count; anti DS-DNA: anti-double stranded DNA antibody.

observed threshold with normative data [pure-tone aver- ordered to rule out retrocochlear pathologies such as
age (PTA) is the arithmetical average of thresholds] and (ii) acoustic neuroma, intracranial metastases, demyelinating
word discrimination, which probes the ability to perceive, or ischemic diseases. AIED is suspected when no clear
process and verbally reproduce phonological units that aetiology is found in the imaging studies, and there is pro-
comprise spoken words. Speech discrimination score/ gression of the documented SNHL that is too slow to be a
word intelligibility score (WIS) is the percentage of words sudden SNHL (which is unilateral and defined as develop-
a patient can repeat at a comfortable listening level. A WIS ing over 72 h), but too fast to be presbycusis (which
of 70% or more is considered indicative of good potential develops over many years).
for spoken communication. So maintaining or reaching a Despite significant efforts, to date there are no reliable
WIS of 70% is a good clinical outcome. Table 2 summar- diagnostic tests for AIED. Lymphocyte migration inhibition
izes the functional impact of different degrees of hearing assays and lymphocyte transformation tests have initially
impairment. been included as part of the diagnostic approach; how-
The next relevant step is to exclude other disorders that ever, their diagnostic accuracy remains undetermined.
can mimic AIED (i.e. multiple sclerosis, malignancies and The use of the enzyme-linked immunospot (ELISPOT)
otosyphilis) [36]. Based on expert opinion, the recom- assay to assess the frequencies of peripheral blood
mended routine serology tests in patients with AIED T cells capable of producing IFN-g in response to hom-
include complete blood count, ESR, RF, ANA test, anti- ogenates of human inner ear tissue in patients with AIED
dsDNA antibodies, anti-SSA/B antibodies, antiphospholi- was tested in a small study including 12 patients. This
pid antibodies, ANCA, C3 and C4 complement levels, study showed that 25% of AIED patients have increased
HIV and fluorescent treponemal antibody absorption test frequencies of inner-ear-specific IFN-g-producing T cells
[30, 44, 45]. In the absence of symptoms and signs of in peripheral blood [23, 49]. Similarly to cellular assays,
autoimmune diseases, the value of including serology serological tests to detect antibodies against inner ear
testing appears limited [46, 47]. Moreover, positive results antigens lack sufficient specificity to be used as diagnos-
of those tests are not predictive of improvement following tic tools [50]. The OtoBlot assay, a commercially available
steroid treatment [39]. Serology studies for viral infections western blot assay for anti-hsp70 antibodies, has low sen-
are not recommended in AIED [48]. sitivity (50%) [29, 47, 51, 52]. More recently, anti-CTL-2
If the cause of the hearing loss is not identified, particu- antibodies were demonstrated in 50% of AIED sera; how-
larly in patients with asymmetric SNHL, an MRI should be ever, this test is not commercially available and its

www.rheumatology.oxfordjournals.org 783
Tamara Mijovic et al.

FIG. 2 Typical audiograms of conductive and sensori- TABLE 2 Scales of hearing impairment and their
neural hearing loss. functional implications based on PTA and speech
discrimination score (SDS)

Functional implication in
Degree of terms of need for hearing
PTA (dB) impairment aid and lipreading

10 to 15 Normal No

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


16–25 Slight Possibly
26–40 Mild Probably
41–65 Moderate Definitely
66–95 Severe Definitely
>95 Profound Definitely

Degree of Functional implication on


SDS (%) impairment conversational abilities

90–100 Normal None


75–90 Mild Slight, comparable to a
conversation over the
phone
60–75 Moderate Moderate difficulty
50–60 Severe Marked difficulty in
following a
conversation
<50 Profound Unable to follow running
speech

Adapted from Semin Arthritis Rheum 2004;34:538–43, with


permission from Elsevier.

capacity occur. A significant change in hearing is defined


by any of the following criteria: a threshold shift of 15 dB or
more in one frequency; a threshold shift of 10 dB or more
at two or more consecutive frequencies; 10 dB or more
change in PTA [37, 56–59]; 12–20% or above change in
discrimination score at 40 dB sensation level [7, 60, 61]. In
addition, to be considered as improvement in hearing,
some authors will request no additional threshold loss at
any other frequency and no additional loss in discrimin-
ation in either of the ears [52].

Steroids
(A) Conductive hearing loss: there is a gap between the Corticosteroid therapy is the mainstay of AIED treatment
hearing thresholds through air and bone. (B) Sensorineural due to its immunosuppressive properties and its effects on
hearing loss: the bone and air conduction curves follow the modulation of sodium transport. There are no prospect-
each other. ive randomized clinical trials on which to base the dose,
route and length of the corticosteroid treatment. Moreover,
diagnostic value needs to be confirmed in larger studies although many patients have short-term response to ster-
[53]. The usefulness of PET for assessing active disease is oids, the response is generally not sustained [62].
controversial [54, 55]. The most widely used regimen by otologists is an empiric
treatment protocol proposed by Rauch et al. [7] based on
Treatment 150 patients in whom autoimmune inner disease was clin-
ically suspected. Initial therapy in adults is a 60 mg/day or
A multidisciplinary team that includes an otolaryngologist, 1 mg/kg/day therapeutic trial of prednisone for 4 weeks.
a rheumatologist and an audiologist should treat a patient Patients’ hearing is tested at the initiation of therapy and
with AIED, monitor therapeutic responses and prevent 1 month later. A response is documented at the end of
side effects associated with immunosuppressants. 4 weeks in 50–70% of cases [38] and those patients con-
Patients with AIED should be followed with audiograms tinue therapy until monthly audiograms demonstrate stabi-
on a monthly basis until their hearing stabilizes, and lization [63]. At that point, steroids are tapered over 8
every 6 months thereafter unless changes in the hearing weeks to a maintenance dose of 10 mg for a total treatment

784 www.rheumatology.oxfordjournals.org
Autoimmune inner ear disease

of at least 6 months. Relapses that are associated with open-label study including 17 patients with refractory
treatment duration of less than 6 months require escal- autoimmune hearing loss, MTX treatment was associated
ation of the dosage until stabilization is achieved. In pa- with hearing improvement in 65% of the cases, stabiliza-
tients who fail to respond to the initial 4-week trial, tion in 23% and worsening in 12% [69]. A discrepancy
steroids are tapered over 12 days. between rates of improvement based on audiometric
Steroid-associated adverse effects occur in 15% of parameters (65%) and on patient’s perception (35%)
AIED patients during the first month of treatment and 6% was evidenced in this study raising the issue of using
have to discontinue steroid therapy [56]. Several strategies the latter as a relevant outcome for future intervention

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


have been proposed to overcome this problem. Some au- studies [69].
thors have suggested the use of shorter steroid regimens In 2003, the only prospective, multicentre, randomized,
[38]; however, those might be associated with higher rate double-blind, placebo-controlled trial in AIED was
of relapse [3]. The use of systemic mineralocorticoids reported. This trial was designed to assess the efficacy
(aldosterone and fludrocortisone) alone or in combination of long-term MTX (52 weeks, MTX dose: 15 mg/week) in
with glucocorticoids seems to be useful in animal models the maintenance of initial hearing improvement in
but has not been tested in humans [64, 65]. Intratympanic response to prednisone therapy in patients with primary
steroid injections have been tested in AIED patients refrac- AIED. The study included 67 patients and was terminated
tory to steroids [66]. Despite high penetration of transtym- early due to no apparent benefit of MTX over placebo in
panic methylprednisolone into the cochlear fluids [67], the maintaining clinical response during prednisone tapering
evidence supporting its use in AIED is limited. In a small [61]. However, this study did not assess the impact of
retrospective case series of 11 patients, transtympanic MTX on vestibular symptoms nor hearing fluctuations
6-methylprednisolone was safe and useful. Those pa- over time, which previous MTX open-label studies re-
tients received weekly injections of 6-methylprednisolone ported to improve [66].
(0.3–0.5 ml, 40 mg MP/ml) for 2 months and 54% of them
had hearing improvement with significant reduction of the Biologics
vestibular symptoms [66]. Some patients prefer to avoid Almost every biologic agent has been used for the treat-
or cannot tolerate the side effects of steroid therapy and ment of AIED. Most of the available evidence on these
in those cases consideration may eventually need to be agents derives from observational studies, case reports
given to cochlear implants if the patient deteriorates to pro- and case series, which can be confounded by selection
found hearing loss. and publication bias. There are no randomized clinical
trials assessing the effectiveness of the use of specific
Non-steroidal immunosuppressants biologics in AIED.
A significant number of patients with AIED have pri-
mary or secondary response failure to steroids, are Cytokine modulation
steroid dependent or experience unacceptable side ef-
fects. In these patients, alternative immunosuppressive Blocking TNF-a, both systemically and locally, has shown
approaches and steroid-sparing agents are required. promising results in models of AIED [70]. In animals, the
Cyclophosphamide appears to be an effective therapy efficacy of etanercept was similar to that of glucocortic-
and its use in association with steroids was first described oids as a primary treatment modality [71]. In humans,
in McCabe’s series [2]. However, the extended follow-up TNF-a blockers have been used in refractory cases
of patients treated with cyclophosphamide for other but not as a first line of therapy. Overall, the avail-
benign conditions has been associated with a high inci- able evidence failed to demonstrate a significant ef-
dence of serious drug-related toxicities prompting fect of anti-TNF agents on hearing outcomes in these
the search for other immunosuppressive options. The patients.
evidence supporting the use of ciclosporin A [57], MMF A retrospective case series of 12 patients with AIED
[37, 58] and AZA [37, 60] in AIED is based on case reports who did not respond to conventional stabilization thera-
and small case series and not conclusive. We will discuss pies but showed initial response to high-dose prednis-
agents for which stronger evidence is available. one, or who experienced side effects with conventional
therapy, were treated with etanercept 25 mg s.c. twice
Methotrexate weekly for 6 months. Improvement or stabilization of
hearing and tinnitus was observed in 92% of the patients
(MTX 7.5–25 mg/week) has been used as a prednisone- [72]. In addition, seven of eight patients who had vertigo
sparing treatment for refractory autoimmune hearing loss (88%) and eight of nine patients who had aural fullness
or as an adjunct to maintain or further improve the symp- (89%) had resolution or significant improvement of their
toms. Non-randomized studies including a prospective symptoms. In contrast to these results, an open-label
observational study of 53 patients demonstrated good tol- prospective pilot study of 23 patients with bilateral im-
erance and an overall 70% response rate with vertigo mune-mediated cochleovestibular disorders treated with
improving in 69%, hearing loss in 53% and tinnitus in etanercept for 24 weeks (25 mg subcutaneously twice
26% [68]. Moreover, in that study, most patients were weekly) did not suggest substantial efficacy, with only
able to stop therapy after 1–2 years without recurrence 30% of patients showing improvement in audiometric
of symptoms [68]. In another prospective 12-month criteria [55]. In this study, hearing did not change in

www.rheumatology.oxfordjournals.org 785
Tamara Mijovic et al.

57% of patients, leaving unanswered the question of Plasmapheresis


whether etanercept might have a role in disease stabi- The benefit of plasmapheresis (PMP) for AIED was evalu-
lization when steroid treatment is inadequate. ated in studies including small number of patients (8–21 pa-
Subsequently, a randomized controlled pilot trial includ- tients). Luetje et al. evaluated the long-term hearing
ing 20 patients with AIED showed that treatment with outcome in 16 patients who had tried other treatments
etanercept 25 mg twice weekly over 8 weeks was no without success and received PMP on alternate days for
better than placebo in improving hearing [73]. The draw- cycles of three exchanges. The average follow-up of these
backs of this study were the small sample size, length of patients was 7 years. The authors suggest that 39% of

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


treatment and the inclusion of a heterogeneous patient the ears met the criteria for hearing improvement or
population with variable disease duration with multiple stability and 25% of the patients required continuing
previous treatments. immunosuppressive drugs [81]. The precise role of PMP
A recent retrospective review of eight patients with in AIED and its relation to immune suppression needs to be
suspected autoimmune hearing loss refractory to all defined.
standard treatments treated with infliximab failed to docu-
ment positive therapeutic responses based on objective
hearing measurements [74]. Since antibodies have been Cochlear implantation
demonstrated to cross the round window membrane [75, The subset of patients with AIED who develop profound
76] and since TNF-a blockers were not found to be oto- irreversible bilateral SNHL who no longer benefit from
toxic, transtympanic infliximab administration was tested hearing aid amplification become candidates for cochlear
[77]. In a prospective pilot study nine patients diagnosed implantation [82]. Outcome studies have demonstrated
with AIED with initial response to oral steroids were trea- excellent results and performance above average in
ted with 0.3 ml of infliximab once weekly for 4 weeks implanted AIED patients, compared with patients with
through a transtympanic tube and a MicroWick. other causes of deafness [82–84].
Response defined as improvement in hearing loss with
steroid tapering and a reduction in the relapse rate was
observed in seven patients [77]. Conclusion
A single case suggesting the benefit of adalimumab
in conjunction with MTX for the control of AIED [59] was The absence of reliable markers for AIED has forced
challenged by two reports of de novo AIED associated clinicians to define the disease based on its clinical pres-
with this monoclonal antibody [78]. entation, exclusion of other known causes, and a positive
A recent study evaluating the pathways that control response to steroid therapy. In every patient who has pro-
corticosteroid responsiveness in 47 patients with AIED gressive bilateral SNHL with no other explainable cause,
has shown that increased levels of IL-1b expression are the diagnosis of AIED should be considered, the associ-
associated with corticosteroid resistance, suggesting a ation with a systemic autoimmune disease evaluated and
role for IL-1b inhibition in clinical corticosteroid non- a trial of corticosteroids attempted.
responders [79]. Rheumatology key messages
. AIED is a steroid-responsive fluctuating progressive
bilateral asymmetrical SNHL that develops over
Cell modulation weeks to months.
. No specific diagnostic tests for AIED exist and
Rituximab (anti-CD20 mAb) was tested in an open-label
15–30% of cases have an associated systemic
study of seven patients with primary immune-mediated autoimmune disease.
inner steroid-responsive AIED. A course of rituximab . Glucocorticoids remain the mainstay of AIED
(1000 mg i.v. for two doses 2 weeks apart) prior to starting treatment.
prednisone tapering was associated with sustained re-
sponse (evaluated at week 24) following steroid discon-
tinuation in five out of seven patients (24-week follow-up)
Disclosure statement: The authors have declared no
[80]. The results of this pilot study support the evaluation
conflicts of interest.
of rituximab in a properly designed randomized clinical
trial [80].
In summary, the current evidence supporting the use of
References
steroid-sparing agents among them biologic agents as a
treatment for AIED is still anecdotal. Properly designed 1 Lehnhardt E. Sudden hearing disorders occurring
randomized controlled trials are necessary to establish simultaneously or successively on both sides. Z Laryngol
the efficacy of biologics as the treatment of AIED. Given Rhinol Otol 1958;37:1–16.
the high cost of these agents, if efficacy is proven, cost- 2 McCabe BF. Autoimmune sensorineural hearing loss. Ann
effectiveness studies comparing them to the alternative of Otol Rhinol Laryngol 1979;88:585–9.
a cochlear implant will certainly be warranted before they 3 Bovo R, Aimoni C, Martini A. Immune-mediated inner ear
become part of the standard treatment. disease. Acta Otolaryngol 2006;126:1012–21.

786 www.rheumatology.oxfordjournals.org
Autoimmune inner ear disease

4 Agrup C, Luxon LM. Immune-mediated inner-ear autoimmune sensorineural hearing loss. J Immunol 2006;
disorders in neuro-otology. Curr Opin Neurol 2006;19: 177:4203–10.
26–32. 23 Lorenz RR, Solares CA, Williams P et al. Interferon-gamma
5 Hughes GB, Kinney SE, Barna BP et al. Practical versus production to inner ear antigens by T cells from patients
theoretical management of autoimmune inner ear disease. with autoimmune sensorineural hearing loss. J
Laryngoscope 1984;94:758–67. Neuroimmunol 2002;130:173–8.
6 Moscicki RA, San Martin JE, Quintero CH et al. Serum 24 Zhou Y, Yuan J, Zhou B et al. The therapeutic efficacy of
antibody to inner ear proteins in patients with progressive human adipose tissue-derived mesenchymal stem cells
on experimental autoimmune hearing loss in mice.

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


hearing loss. Correlation with disease activity and
response to corticosteroid treatment. JAMA 1994;272: Immunology 2011;133:133–40.
611–6. 25 Helfgott SM, Mosciscki RA, San Martin J et al. Correlation
7 Rauch SD. Clinical management of immune-mediated between antibodies to type II collagen and treatment
inner-ear disease. Ann NY Acad Sci 1997;830:203–10. outcome in bilateral progressive sensorineural hearing
loss. Lancet 1991;337:387–9.
8 Greco A, Fusconi M, Gallo A et al. Sudden sensorineural
hearing loss: an autoimmune disease? Autoimmun Rev 26 Boulassel MR, Tomasi JP, Deggouj N et al. Identification
2011;10:756–61. of beta-actin as a candidate autoantigen in autoimmune
inner ear disease. Clin Otolaryngol Allied Sci 2000;25:
9 Harris JP. Immunology of the inner ear: response of the
535–41.
inner ear to antigen challenge. Otolaryngol Head Neck
Surg 1983;91:18–32. 27 Harris JP, Sharp PA. Inner ear autoantibodies in patients
with rapidly progressive sensorineural hearing loss.
10 Harris JP, Woolf NK, Ryan AF. Elaboration of systemic
Laryngoscope 1990;100:516–24.
immunity following inner ear immunization. Am J
Otolaryngol 1985;6:148–52. 28 Trune DR, Kempton JB, Mitchell CR et al. Failure of
elevated heat shock protein 70 antibodies to alter cochlear
11 Ma C, Billings P, Harris JP et al. Characterization of an
function in mice. Hear Res 1998;116:65–70.
experimentally induced inner ear immune response.
Laryngoscope 2000;110:451–6. 29 Yeom K, Gray J, Nair TS et al. Antibodies to HSP-70 in
normal donors and autoimmune hearing loss patients.
12 Buniel MC, Geelan-Hansen K, Weber PC et al.
Laryngoscope 2003;113:1770–6.
Immunosuppressive therapy for autoimmune inner ear
disease. Immunotherapy 2009;1:425–34. 30 Hirose K, Wener MH, Duckert LG. Utility of laboratory
testing in autoimmune inner ear disease. Laryngoscope
13 Tomiyama S, Harris JP. The endolymphatic sac: its
1999;109:1749–54.
importance in inner ear immune responses. Laryngoscope
1986;96:685–91. 31 Nair TS, Kozma KE, Hoefling NL et al. Identification and
characterization of choline transporter-like protein 2, an
14 Tomiyama S, Harris JP. The role of the endolymphatic sac
inner ear glycoprotein of 68 and 72 kDa that is the target of
in inner ear immunity. Acta Otolaryngol 1987;103:182–8. antibody-induced hearing loss. J Neurosci 2004;24:
15 Yeo SW, Gottschlich S, Harris JP et al. Antigen diffusion 1772–9.
from the perilymphatic space of the cochlea. 32 Harris JP, Heydt J, Keithley EM et al. Immunopathology of
Laryngoscope 1995;105:623–8. the inner ear: an update. Ann NY Acad Sci 1997;830:
16 Satoh H, Firestein GS, Billings PB et al. Proinflammatory 166–78.
cytokine expression in the endolymphatic sac during 33 Hoistad DL, Schachern PA, Paparella MM. Autoimmune
inner ear inflammation. J Assoc Res Otolaryngol 2003;4: sensorineural hearing loss: a human temporal bone study.
139–47. Am J Otolaryngol 1998;19:33–9.
17 Satoh H, Billings P, Firestein GS et al. Transforming 34 Orozco CR, Niparko JK, Richardson BC et al.
growth factor beta expression during an inner ear immune Experimental model of immune-mediated hearing loss
response. Ann Otol Rhinol Laryngol 2006;115:81–8. using cross-species immunization. Laryngoscope 1990;
18 Gloddek B, Gloddek J, Arnold W. Induction of an 100:941–7.
inner-ear-specific autoreactive T-cell line for the diagnos- 35 Eisen MD, Niparko JK. Autoimmune inner ear disease.
tic evaluation of an autoimmune disease of the inner ear. Eggers SDZ, Zee DS, eds. Vertigo and Imbalance: Clinical
Ann N Y Acad Sci 1997;830:266–76. Neurophysiology of the Vestibular System. Handbook of
19 Ikezono T, Tomiyama S, Pawankar R et al. Passive Clinical Neurophysiology, Vol. 9. Amsterdam: Elsevier BV,
transfer of experimental autoimmune labyrinthitis. Audiol 2010:28–432.
Neurootol 2000;5:292–9. 36 George DL, Pradhan S. Idiopathic sensorineural hearing
20 Ikezono T, Omori A, Ichinose S et al. Identification of the disorders in adults—a pragmatic approach. Nat Rev
protein product of the coch gene (hereditary deafness Rheumatol 2009;5:505–12.
gene) as the major component of bovine inner ear protein. 37 Broughton SS, Meyerhoff WE, Cohen SB.
Biochim Biophys Acta 2001;1535:258–65. Immune-mediated inner ear disease: 10-year experience.
21 Solares CA, Edling AE, Johnson JM et al. Murine autoim- Semin Arthritis Rheum 2004;34:544–8.
mune hearing loss mediated by CD4+ T cells specific for 38 Loveman DM, de Comarmond C, Cepero R et al.
inner ear peptides. J Clin Invest 2004;113:1210–7. Autoimmune sensorineural hearing loss: clinical course
22 Baek MJ, Park HM, Johnson JM et al. Increased and treatment outcome. Semin Arthritis Rheum 2004;34:
frequencies of cochlin-specific T cells in patients with 538–43.

www.rheumatology.oxfordjournals.org 787
Tamara Mijovic et al.

39 Hervier B, Bordure P, Audrain M et al. Systematic multi-center, open-label, pilot study. Arthritis Rheum 2005;
screening for nonspecific autoantibodies in idiopathic 53:337–42.
sensorineural hearing loss: no association with steroid 56 Alexander TH, Weisman MH, Derebery JM et al. Safety
response. Otol Neurotol 2010;31:687–90. of high-dose corticosteroids for the treatment of
40 Gazquez I, Soto-Varela A, Aran I et al. High prevalence of autoimmune inner ear disease. Otol Neurotol 2009;30:
systemic autoimmune diseases in patients with Meniere’s 443–8.
disease. PLoS One 2011;6:e26759. 57 Di Leo E, Coppola F, Nettis E et al. Late recovery with
41 Bakthavachalam S, Driver MS, Cox C et al. Hearing cyclosporine-A of an auto-immune sudden sensori-
neural hearing loss. Acta Otorhinolaryngol Ital 2011;31:

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


loss in Wegener’s granulomatosis. Otol Neurotol 2004;25:
833–7. 399–401.
42 Bachor E, Blevins NH, Karmody C et al. Otologic 58 Hautefort C, Loundon N, Montchilova M et al.
manifestations of relapsing polychondritis. Review of Mycophenolate mofetil as a treatment of steroid
literature and report of nine cases. Auris Nasus Larynx dependent Cogan’s syndrome in childhood. Int J Pediatr
2006;33:135–41. Otorhinolaryngol 2009;73:1477–9.
43 Halligan CS, Bauch CD, Brey RH et al. Hearing loss in 59 Morovic Vergles J, Radic M, Kovacic J et al. Successful
rheumatoid arthritis. Laryngoscope 2006;116:2044–9. use of adalimumab for treating rheumatoid arthritis with
autoimmune sensorineural hearing loss: two birds with
44 Rauch SD, Cohen MA, Ruckenstein MJ. Autoimmune
one stone. J Rheumatol 2010;37:1080–1.
inner ear disease. In: Flint PW, ed. Cummings
Otolaryngology Head and Neck Surgery. Philadelphia, PA: 60 Saracaydin A, Katircioglu S, Karatay MC. Azathioprine in
Mosby Elsevier, 2010:2164–8. combination with steroids in the treatment of autoimmune
inner-ear disease. J Int Med Res 1993;21:192–6.
45 Garcia Callejo FJ, Marco Algarra J, Martinez Beneyto MP
et al. Autoimmune identification of sudden hearing loss. 61 Harris JP, Weisman MH, Derebery JM et al. Treatment of
Acta Otolaryngol 2003;123:168–71. corticosteroid-responsive autoimmune inner ear disease
with methotrexate: a randomized controlled trial. JAMA
46 Garcia Callejo FJ, Corts J, de Paula Vernetta C et al.
2003;290:1875–83.
[Cormobidity of sensorineural hearing loss and other
autoimmune diseases. Usefulness of laboratory tests]. 62 Zeitoun H, Beckman JG, Arts HA et al. Corticosteroid
Acta Otorrinolaringol Esp 2006;57:28–33. response and supporting cell antibody in autoimmune
hearing loss. Arch Otolaryngol Head Neck Surg 2005;131:
47 Garcia-Berrocal JR, Trinidad A, Ramirez-Camacho R et al.
665–72.
Immunologic work-up study for inner ear disorders:
looking for a rational strategy. Acta Otolaryngol 2005;125: 63 Niparko JK, Wang NY, Rauch SD et al. Serial audiometry
814–8. in a clinical trial of AIED treatment. Otol Neurotol 2005;26:
908–17.
48 Garcia-Berrocal JR, Ramirez-Camacho R,
Gonzalez-Garcia JA et al. Does the serological study for 64 MacArthur CJ, Kempton JB, DeGagne J et al. Control of
viral infection in autoimmune inner ear disease make chronic otitis media and sensorineural hearing loss in
sense? ORL J Otorhinolaryngol Relat Spec 2008;70:16–9; C3H/HeJ mice: glucocorticoids vs mineralocorticoids.
discussion 19–20. Otolaryngol Head Neck Surg 2008;139:646–53.
49 Solares CA, Tuohy VK. ELISPOT determination of 65 Trune DR. Ion homeostasis in the ear: mechanisms,
interferon-gamma T-cell frequencies in patients with maladies, and management. Curr Opin Otolaryngol Head
autoimmune sensorineural hearing loss. Methods Mol Biol Neck Surg 2010;18:413–9.
2005;302:253–60. 66 Garcia-Berrocal JR, Ibanez A, Rodriguez A et al.
50 Pham BN, Rudic M, Bouccara D et al. Antibodies to myelin Alternatives to systemic steroid therapy for refrac-
protein zero (p0) protein as markers of auto-immune inner tory immune-mediated inner ear disease: a physiopatho-
ear diseases. Autoimmunity 2007;40:202–7. logic approach. Eur Arch Otorhinolaryngol 2006;263:
51 Garcia Berrocal JR, Ramirez-Camacho R, Arellano B et al. 977–82.
Validity of the western blot immunoassay for heat shock 67 Parnes LS, Sun AH, Freeman DJ. Corticosteroid
protein-70 in associated and isolated immunorelated inner pharmacokinetics in the inner ear fluids: an animal
ear disease. Laryngoscope 2002;112:304–9. study followed by clinical application. Laryngoscope 1999;
52 Bonaguri C, Orsoni JG, Zavota L et al. Anti-68 kDa 109:1–17.
antibodies in autoimmune sensorineural hearing loss: are 68 Salley LH Jr, Grimm M, Sismanis A et al. Methotrexate in
these autoantibodies really a diagnostic tool? the management of immune mediated cochleovestibular
Autoimmunity 2007;40:73–8. disorders: clinical experience with 53 patients.
53 Kommareddi PK, Nair TS, Vallurupalli M et al. J Rheumatol 2001;28:1037–40.
Autoantibodies to recombinant human CTL2 in 69 Matteson EL, Fabry DA, Facer GW et al. Open trial of
autoimmune hearing loss. Laryngoscope 2009;119: methotrexate as treatment for autoimmune hearing loss.
924–32. Arthritis Rheum 2001;45:146–50.
54 Mazlumzadeh M, Lowe VJ, Mullan BP et al. The utility of 70 Wang X, Truong T, Billings PB et al. Blockage of
positron emission tomography in the evaluation of immune-mediated inner ear damage by etanercept. Otol
autoimmune hearing loss. Otol Neurotol 2003;24:201–4. Neurotol 2003;24:52–7.
55 Matteson EL, Choi HK, Poe DS et al. Etanercept therapy 71 Lobo D, Trinidad A, Garcia-Berrocal JR et al. TNFalpha
for immune-mediated cochleovestibular disorders: a blockers do not improve the hearing recovery obtained

788 www.rheumatology.oxfordjournals.org
Autoimmune inner ear disease

with glucocorticoid therapy in an autoimmune 78 Conway R, Khan S, Foley-Nolan D. Use of adalimumab in


experimental labyrinthitis. Eur Arch Otorhinolaryngol 2006; treatment of autoimmune sensorineural hearing loss: a
263:622–6. word of caution. J Rheumatol 2011;38:176.
72 Rahman MU, Poe DS, Choi HK. Etanercept therapy 79 Pathak S, Goldofsky E, Vivas EX et al. IL-1beta is
for immune-mediated cochleovestibular disorders: overexpressed and aberrantly regulated in corticosteroid
preliminary results in a pilot study. Otol Neurotol 2001;22: nonresponders with autoimmune inner ear disease.
619–24. J Immunol 2011;186:1870–9.
73 Cohen S, Shoup A, Weisman MH et al. Etanercept 80 Cohen S, Roland P, Shoup A et al. A pilot study of

Downloaded from https://academic.oup.com/rheumatology/article/52/5/780/1816947 by University of South Florida user on 06 October 2020


treatment for autoimmune inner ear disease: results of a rituximab in immune-mediated inner ear disease. Audiol
pilot placebo-controlled study. Otol Neurotol 2005;26: Neurootol 2011;16:214–21.
903–7.
81 Luetje CM, Berliner KI. Plasmapheresis in autoimmune
74 Liu YC, Rubin R, Sataloff RT. Treatment-refractory inner ear disease: long-term follow-up. Am J Otol 1997;18:
autoimmune sensorineural hearing loss: response to 572–6.
infliximab. Ear Nose Throat J 2011;90:23–8.
82 Aftab S, Semaan MT, Murray GS et al. Cochlear
75 Harris JP, Ryan AF. Effect of a middle ear immune implantation outcomes in patients with autoimmune and
response on inner ear antibody levels. Ann Otol Rhinol immune-mediated inner ear disease. Otol Neurotol 2010;
Laryngol 1985;94:202–6. 31:1337–42.
76 Keithley EM, Ryan AF, Harris JP. Role of the round 83 Wang NM, Wu CM, Kirk KI. Lexical effects on spoken
window membrane in middle ear immune responses. Acta word recognition performance among Mandarin-speaking
Otolaryngol Suppl 1989;457:139–43. children with normal hearing and cochlear implants. Int J
77 Van Wijk F, Staecker H, Keithley E et al. Local perfusion of Pediatr Otorhinolaryngol 2010;74:883–90.
the tumor necrosis factor alpha blocker infliximab to the 84 Benson AG. Labyrinthitis ossificans secondary to
inner ear improves autoimmune neurosensory hearing autoimmune inner ear disease: a previously unreported
loss. Audiol Neurootol 2006;11:357–65. condition. Otolaryngol Head Neck Surg 2010;142:772–3.

www.rheumatology.oxfordjournals.org 789

You might also like