Autoimmune Disease of the Inner Ear

Back

Practice Essentials

In 1979, McCabe first described a cohort of patients with idiopathic, rapidly progressive bilateral sensorineural hearing loss (SNHL). These patients' hearing improved after treatment with corticosteroids, thereby suggesting an autoimmune pathogenesis. The hallmark of this clinically diagnosed condition is the presence of a rapidly progressive, often fluctuating, bilateral SNHL over a period of weeks to months. The progression of hearing loss is too rapid to be diagnostic for presbycusis and too slow to conclude a diagnosis of sudden SNHL. Vestibular symptoms, such as true vertigo, generalized imbalance, and ataxia, may be present.[1]

See the image below.



View Image

Inner ear.

Antigen-nonspecific tests are useful in routine screening for evidence of systemic immunologic dysfunction, yet specifically are not known to correlate with a diagnosis of immune-mediated inner ear disease.

Antigen-specific tests are as follows:

The natural history of untreated immune-mediated inner ear disease is unknown; much of the current therapy is based on empiric clinical data gathered during the past few decades. A key feature of immune-mediated inner ear disease is a positive response to immunosuppressive therapy (ie, corticosteroids) in the form of improved hearing.[2]

Surgery generally is not appropriate for immune-mediated inner ear disease. However, intratympanic therapy performed under local anesthesia has been found to be beneficial for some patients.

Pathophysiology

The term autoimmune inner ear disease (AIED) implies a direct attack of the immune system upon an endogenous inner ear antigen. Most of the evidence linking the immune system to cochleovestibular dysfunction is indirect; therefore, immune-mediated inner ear disease may be a preferred term. AIED is a clinical diagnosis based on its distinct clinical course, immune test results, and treatment response. The most important diagnostic finding is improvement in hearing observed with a trial of immunosuppressants.[3, 4, 5, 6]

Specific criteria for idiopathic progressive bilateral sensorineural hearing loss (IPBSNHL) include bilateral SNHL of at least 30 dB at any frequency with progression in at least one ear, defined as a threshold shift that is greater than 15 dB at any frequency or 10 dB at 2 or more consecutive frequencies or a significant change in discrimination score. This definition excludes patients with sudden SNHL occurring in less than 24 hours, which more likely is due to a microvascular or viral etiology.

A certain subset of patients with presumed Ménière disease (idiopathic endolymphatic hydrops) actually may have Ménière syndrome, in which the underlying pathophysiology is immune mediated. Typically, Ménière disease is initially diagnosed in these patients; however, fluctuating hearing loss in the contralateral ear develops later. This change may prompt a workup for AIED. Hughes et al found that approximately one half of their patients with AIED have manifestations of autoimmune Ménière syndrome.[7, 8]

Epidemiology

Frequency

United States

Because the existence of autoimmune inner ear disease (AIED) has been recognized only since 1979, incidence is difficult to determine. Recent studies in the literature from large referral centers are based on relatively small sample sizes of patients who fit the criteria for diagnosis of AIED. As diagnostic tests for the condition become more specific and more is known about AIED, more patients will be identified who have an autoimmune basis for inner ear symptoms.

Sex

The condition has been suggested to be more common in female patients who may or may not have concomitant systemic autoimmune disease than in male patients.

Age

In most patients, initial onset of symptoms occurs at age 20-50 years. Cases in pediatric patients are uncommon.[9]

History

See the list below:

Physical

Findings from physical examination of the ear usually are normal in patients with immune-mediated inner ear disease. Occasionally, associated systemic autoimmune diseases can affect the external ear skin or middle ear mucosa.

Causes

See the list below:

Laboratory Studies

Antigen-nonspecific tests are useful in routine screening for evidence of systemic immunologic dysfunction, yet specifically are not known to correlate with a diagnosis of immune-mediated inner ear disease. Antigen-nonspecific tests are as follows:

A study by Dayal et al discovered that patients with autoimmune ear disease having no systemic autoimmune illness often show high levels of antinuclear antibodies (with a speckled pattern) and also sometimes have high levels of rheumatoid factor. The positive yield of other detailed tests was low.[15]

A study by Svrakic et al indicated that tumor necrosis factor (TNF) levels can be used diagnostically and prognostically in immune-mediated inner ear disease. The study, which involved 85 patients with clinical and audiometric characteristics of immune-mediated SNHL, as well as 11 controls, found that patients with immune-mediated SNHL who were nonresponsive to steroids had a higher mean baseline plasma level of TNF (27.6 pg/mL), as derived from peripheral venous blood, than did those who were responsive to steroids (24.1 pg/mL), and that both had higher TNF levels than did the controls (14.4 pg/mL). According to the investigators, a baseline plasma TNF level of more than 18.8 pg/mL from the peripheral circulation has a positive predictive value for immune-mediated SNHL of more than 97%.[16]

Svrakic and colleagues also found evidence that in non-steroid-responsive patients with immune-mediated SNHL, peripheral blood mononuclear cells (PBMCs) respond differently to in vitro stimulation with dexamethasone than do those of steroid-responsive patients, with the mean TNF level secreted by the PBMCs slightly increasing (from 11.2 pg/mL to 11.7 pg/mL) with stimulation instead of decreasing.[16]

Antigen-specific tests are as follows:

Histologic Findings

Histopathologic human temporal bone studies of patients with immune-mediated inner ear disease rarely are reported in the literature. Further studies may help elucidate the pathophysiology involved in this condition.

Medical Care

The natural history of untreated immune-mediated inner ear disease is unknown; much of the current therapy is based on empiric clinical data gathered during the past few decades. A key feature of immune-mediated inner ear disease is a positive response to immunosuppressive therapy (ie, corticosteroids) in the form of improved hearing.[2]

Surgical Care

Immune-mediated inner ear disease usually is bilateral and often responds to medical management. Surgery generally is not appropriate. However, intratympanic therapy performed under local anesthesia has been found to be beneficial for some patients with immune-mediated inner ear disease.

As treatment options improve for many inner ear diseases and injuries, methods for delivering precise and controlled doses become vital. Researchers in the field of inner ear drug delivery are constantly in the process of advancing new and existing techniques that support the arrival of better and better therapeutic compounds. Those suffering from hearing related disorders can look forward to improved quality of life as the field progresses.[31]

However, physicians must realize that a potential impact of glucocorticoids on ion homeostasis functions exists in addition to immune suppression. These functions are quite interlinked with regard to maintenance of the endolymphatic potential in fluids around auditory and vestibular hair cells. Therefore, assuming that all steroid-responsive hearing loss is due to immune processes simply cannot be justified in light of our current understanding of other cellular and molecular processes under the control of glucocorticoids.[32]

In a retrospective study of intratympanic dexamethasone for sudden sensorineural hearing loss after failure of systemic therapy, 39% of patients were found to recover 20 dB or 20% SDS (if treated within 6 weeks). This was higher than the figure of 9.1% in their controls.[33]

Consultations

Consultation with a rheumatologist or hematologist often is necessary if steroid therapy fails to help the patient and he or she requires treatment with cytotoxic drugs, which requires close hematologic monitoring. Occasionally, patients may require treatment with plasmapheresis (see Medical Care).

Diet

Patients with immune-mediated endolymphatic hydrops usually are placed on a low-sodium diet, similar to the diet recommended for patients with Ménière disease.

Medication Summary

The goals of pharmacotherapy are to prevent complications and reduce morbidity.

Prednisone (Sterapred)

Clinical Context:  Immunosuppressant for treatment of autoimmune disorders; may decrease inflammation by reversing increased capillary permeability and suppressing PMN activity.

Class Summary

These agents have anti-inflammatory properties and cause profound and varied metabolic effects. Corticosteroids modify the body's immune response to diverse stimuli.

Cyclophosphamide (Cytoxan, Neosar)

Clinical Context:  Chemically related to nitrogen mustards. An alkylating agent; mechanism of action of active metabolites may involve cross-linking of DNA, which may interfere with growth of normal and neoplastic cells.

Methotrexate (Folex PFS, Rheumatrex)

Clinical Context:  Unknown mechanism of action in treatment of inflammatory reactions; may affect immune function. Ameliorates symptoms of inflammation (eg, pain, swelling, stiffness).

Adjust dose gradually to attain satisfactory response.

Class Summary

These agents inhibit cell growth and proliferation and are useful in the treatment of autoimmune diseases.

Prognosis

See the list below:

Patient Education

See the list below:

Author

Neeraj N Mathur, MBBS, MS, DNB(ENT), MNAMS, FAMS, Principal and Director-Professor (ENT), Vardhman Mahavir Medical College and Safdarjung Hospital; Professor, Guru Gobind Singh Indraprastha University and Delhi University, India

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Peter S Roland, MD, Professor, Department of Neurological Surgery, Professor and Chairman, Department of Otolaryngology-Head and Neck Surgery, Director, Clinical Center for Auditory, Vestibular, and Facial Nerve Disorders, Chief of Pediatric Otology, University of Texas Southwestern Medical Center; Chief of Pediatric Otology, Children’s Medical Center of Dallas; President of Medical Staff, Parkland Memorial Hospital; Adjunct Professor of Communicative Disorders, School of Behavioral and Brain Sciences, Chief of Medical Service, Callier Center for Communicative Disorders, University of Texas School of Human Development

Disclosure: Received honoraria from Alcon Labs for consulting; Received honoraria from Advanced Bionics for board membership; Received honoraria from Cochlear Corp for board membership; Received travel grants from Med El Corp for consulting.

Chief Editor

Arlen D Meyers, MD, MBA, Professor of Otolaryngology, Dentistry, and Engineering, University of Colorado School of Medicine

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Cerescan;RxRevu;Cliexa;The Physicians Edge;Sync-n-Scale;mCharts<br/>Received income in an amount equal to or greater than $250 from: The Physicians Edge, Cliexa<br/> Received stock from RxRevu; Received ownership interest from Cerescan for consulting; .

Additional Contributors

Robert A Battista, MD, FACS, Assistant Professor of Otolaryngology, Northwestern University, The Feinberg School of Medicine; Physician, Ear Institute of Chicago, LLC

Disclosure: Nothing to disclose.

Acknowledgements

The authors and editors of Medscape Drugs & Diseases gratefully acknowledge the contributions of previous authors Shelley Jaquish, MD, and William L Meyerhoff, MD, PhD, to the development and writing of this article.

References

  1. Vambutas A, Pathak S. AAO: Autoimmune and Autoinflammatory (Disease) in Otology: What is New in Immune-Mediated Hearing Loss. Laryngoscope Investig Otolaryngol. 2016 Oct. 1 (5):110-115. [View Abstract]
  2. Pathak S, Goldofsky E, Vivas EX, Bonagura VR, Vambutas A. IL-1ß is overexpressed and aberrantly regulated in corticosteroid nonresponders with autoimmune inner ear disease. J Immunol. 2011 Feb 1. 186(3):1870-9. [View Abstract]
  3. Goodall AF, Siddiq MA. Current understanding of the pathogenesis of autoimmune inner ear disease: a review. Clin Otolaryngol. 2015 Oct. 40 (5):412-9. [View Abstract]
  4. Lobo DR, García-Berrocal JR, Ramírez-Camacho R. New prospects in the diagnosis and treatment of immune-mediated inner ear disease. World J Methodol. 2014 Jun 26. 4 (2):91-8. [View Abstract]
  5. Ralli M, D'Aguanno V, Di Stadio A, et al. Audiovestibular Symptoms in Systemic Autoimmune Diseases. J Immunol Res. 2018. 2018:5798103. [View Abstract]
  6. Li G, You D, Ma J, Li W, Li H, Sun S. The Role of Autoimmunity in the Pathogenesis of Sudden Sensorineural Hearing Loss. Neural Plast. 2018. 2018:7691473. [View Abstract]
  7. Hughes GB, Barna BP, Calarese LH. Immunologic Disorders of the Inner Ear. Bailey BJ, ed. Head and Neck Surgery-Otolaryngology. Philadelphia, Pa: Lippincott; 1993. 1833-1842.
  8. Berlinger NT. Meniere's disease: new concepts, new treatments. Minn Med. 2011 Nov. 94(11):33-6. [View Abstract]
  9. Huang NC, Sataloff RT. Autoimmune inner ear disease in children. Otol Neurotol. 2011 Feb. 32(2):213-6. [View Abstract]
  10. Yoo TJ, Tomoda K, Stuart JM, Cremer MA, Townes AS, Kang AH. Type II collagen-induced autoimmune sensorineural hearing loss and vestibular dysfunction in rats. Ann Otol Rhinol Laryngol. 1983 May-Jun. 92(3 Pt 1):267-71. [View Abstract]
  11. Harris JP, Sharp PA. Inner ear autoantibodies in patients with rapidly progressive sensorineural hearing loss. Laryngoscope. 1990 May. 100(5):516-24. [View Abstract]
  12. Suchan M, Kaliarik L, Krempaska S, Koval J. Immune-mediated cochleovestibular disease. Bratisl Lek Listy. 2016. 117 (2):87-90. [View Abstract]
  13. Shamriz O, Tal Y, Gross M. Autoimmune Inner Ear Disease: Immune Biomarkers, Audiovestibular Aspects, and Therapeutic Modalities of Cogan's Syndrome. J Immunol Res. 2018. 2018:1498640. [View Abstract]
  14. D'Aguanno V, Ralli M, de Vincentiis M, Greco A. Optimal management of Cogan's syndrome: a multidisciplinary approach. J Multidiscip Healthc. 2018. 11:1-11. [View Abstract]
  15. Dayal VS, Ellman M, Sweiss N. Autoimmune inner ear disease: clinical and laboratory findings and treatment outcome. J Otolaryngol Head Neck Surg. 2008 Aug. 37(4):591-6. [View Abstract]
  16. Svrakic M, Pathak S, Goldofsky E, et al. Diagnostic and prognostic utility of measuring tumor necrosis factor in the peripheral circulation of patients with immune-mediated sensorineural hearing loss. Arch Otolaryngol Head Neck Surg. 2012 Nov. 138(11):1052-8. [View Abstract]
  17. Moscicki RA, San Martin JE, Quintero CH, Rauch SD, Nadol JB Jr, Bloch KJ. Serum antibody to inner ear proteins in patients with progressive hearing loss. Correlation with disease activity and response to corticosteroid treatment. JAMA. 1994 Aug 24-31. 272(8):611-6. [View Abstract]
  18. Rauch SD, San Martin JE, Moscicki RA, Bloch KJ. Serum antibodies against heat shock protein 70 in Menière's disease. Am J Otol. 1995 Sep. 16(5):648-52. [View Abstract]
  19. Gong SS, Yu DZ, Wang JB. Relationship between three inner ear antigens with different molecular weights and autoimmune inner ear disease. Acta Otolaryngol. 2002 Jan. 122(1):5-9. [View Abstract]
  20. Cao MY, Deggouj N, Gersdorff M, Tomasi JP. Guinea pig inner ear antigens: extraction and application to the study of human autoimmune inner ear disease. Laryngoscope. 1996 Feb. 106(2 Pt 1):207-12. [View Abstract]
  21. McCabe BF. Autoimmune sensorineural hearing loss. Ann Otol Rhinol Laryngol. 1979 Sep-Oct. 88(5 Pt 1):585-9. [View Abstract]
  22. Sismanis A, Thompson T, Willis HE. Methotrexate therapy for autoimmune hearing loss: a preliminary report. Laryngoscope. 1994 Aug. 104(8 Pt 1):932-4. [View Abstract]
  23. Mata-Castro N, Gavilanes-Plasencia J, Ramirez-Camacho R, Garcia-Fernandez A, Garcia-Berrocal JR. Azathioprine reduces the risk of audiometric relapse in immune-mediated hearing loss. Acta Otorrinolaringol Esp. 2018 Sep - Oct. 69 (5):260-7. [View Abstract]
  24. Harris JP, Weisman MH, Derebery JM, et al. Treatment of corticosteroid-responsive autoimmune inner ear disease with methotrexate: a randomized controlled trial. JAMA. 2003 Oct 8. 290(14):1875-83. [View Abstract]
  25. Cohen S, Shoup A, Weisman MH, Harris J. Etanercept treatment for autoimmune inner ear disease: results of a pilot placebo-controlled study. Otol Neurotol. 2005 Sep. 26(5):903-7. [View Abstract]
  26. Matteson EL, Choi HK, Poe DS, et al. Etanercept therapy for immune-mediated cochleovestibular disorders: a multi-center, open-label, pilot study. Arthritis Rheum. 2005 Jun 15. 53(3):337-42. [View Abstract]
  27. Pathak S, Stern C, Vambutas A. N-Acetylcysteine attenuates tumor necrosis factor alpha levels in autoimmune inner ear disease patients. Immunol Res. 2015 Dec. 63 (1-3):236-45. [View Abstract]
  28. Luetje CM. Theoretical and practical implications for plasmapheresis in autoimmune inner ear disease. Laryngoscope. 1989 Nov. 99(11):1137-46. [View Abstract]
  29. Parnes LS, Sun AH, Freeman DJ. Corticosteroid pharmacokinetics in the inner ear fluids: an animal study followed by clinical application. Laryngoscope. 1999 Jul. 109(7 Pt 2):1-17. [View Abstract]
  30. Silverstein H. Use of a new device, the MicroWick, to deliver medication to the inner ear. Ear Nose Throat J. 1999 Aug. 78(8):595-8, 600. [View Abstract]
  31. Swan EE, Mescher MJ, Sewell WF, Tao SL, Borenstein JT. Inner ear drug delivery for auditory applications. Adv Drug Deliv Rev. 2008 Dec 14. 60(15):1583-99. [View Abstract]
  32. Hamid M, Trune D. Issues, indications, and controversies regarding intratympanic steroid perfusion. Curr Opin Otolaryngol Head Neck Surg. 2008 Oct. 16(5):434-40. [View Abstract]
  33. Haynes DS, O'Malley M, Cohen S, Watford K, Labadie RF. Intratympanic dexamethasone for sudden sensorineural hearing loss after failure of systemic therapy. Laryngoscope. 2007 Jan. 117(1):3-15. [View Abstract]

Inner ear.

Inner ear.