CC BY-NC-ND 4.0 · J Am Acad Audiol 2021; 32(10): 661-669
DOI: 10.1055/s-0042-1750281
Review Article
Special Issue on Hearing Therapeutics and Protective Therapies

Approaches to Treat Sensorineural Hearing Loss by Hair-Cell Regeneration: The Current State of Therapeutic Developments and Their Potential Impact on Audiological Clinical Practice

Ashley S. Hinton
1   Frequency Therapeutics, Lexington, Massachusetts
,
Aizhen Yang-Hood
2   Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, Missouri
,
Angela D. Schrader
2   Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, Missouri
,
Christopher Loose
1   Frequency Therapeutics, Lexington, Massachusetts
,
Kevin K. Ohlemiller
2   Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, Missouri
,
1   Frequency Therapeutics, Lexington, Massachusetts
3   Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut
› Author Affiliations

Abstract

Sensorineural hearing loss (SNHL) is typically a permanent and often progressive condition that is commonly attributed to sensory cell loss. All vertebrates except mammals can regenerate lost sensory cells. Thus, SNHL is currently only treated with hearing aids or cochlear implants. There has been extensive research to understand how regeneration occurs in nonmammals, how hair cells form during development, and what limits regeneration in maturing mammals. These studies motivated efforts to identify therapeutic interventions to regenerate hair cells as a treatment for hearing loss, with a focus on targeting supporting cells to form new sensory hair cells. The approaches include gene therapy and small molecule delivery to the inner ear. At the time of this publication, early-stage clinical trials have been conducted to test targets that have shown evidence of regenerating sensory hair cells in preclinical models. As these potential treatments move closer to a clinical reality, it will be important to understand which therapeutic option is most appropriate for a given population. It is also important to consider which audiological tests should be administered to identify hearing improvement while considering the pharmacokinetics and mechanism of a given approach. Some impacts on audiological practice could include implementing less common audiological measures as standard procedure. As devices are not capable of repairing the damaged underlying biology, hair-cell regeneration treatments could allow patients to benefit more from their devices, move from a cochlear implant candidate to a hearing aid candidate, or move a subject to not needing an assistive device. Here, we describe the background, current state, and future implications of hair-cell regeneration research.

Disclaimer

Any mention of a product, service, or procedure in the Journal of the American Academy of Audiology does not constitute an endorsement of the product, service, or procedure by the American Academy of Audiology.




Publication History

Received: 13 March 2021

Accepted: 15 July 2021

Article published online:
24 May 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Blackwell DL, Lucas JW, Clarke TC. Summary health statistics for U.S. adults: national health interview survey, 2012. Vital Heal Stat 10 2014; (260) 1-161
  • 2 Lin FR, Niparko JK, Ferrucci L. Hearing loss prevalence in the United States. Arch Intern Med 2011; 171 (20) 1851-1852
  • 3 Cunningham LL, Tucci DL. Hearing loss in adults. N Engl J Med 2017; 377 (25) 2465-2473
  • 4 World Health Organization (WHO). Hearing loss due to recreational exposure to loud sounds: a review. 2015
  • 5 Li L, Chao T, Brant J, O'Malley Jr B, Tsourkas A, Li D. Advances in nano-based inner ear delivery systems for the treatment of sensorineural hearing loss. Adv Drug Deliv Rev 2017; 108: 2-12
  • 6 Basner M, Brink M, Bristow A. et al. ICBEN review of research on the biological effects of noise 2011-2014. Noise Health 2015; 17 (75) 57-82
  • 7 Goman AM, Reed NS, Lin FR. Addressing estimated hearing loss in adults in 2060. JAMA Otolaryngol Head Neck Surg 2017; 143 (07) 733-734
  • 8 Wu PZ, O'Malley JT, de Gruttola V, Liberman MC. Age-related hearing loss is dominated by damage to inner ear sensory cells, not the cellular battery that powers them. J Neurosci 2020; 40 (33) 6357-6366
  • 9 Choi JS, Betz J, Li L. et al. Association of using hearing aids or cochlear implants with changes in depressive symptoms in older adults. JAMA Otolaryngol Head Neck Surg 2016; 142 (07) 652-657
  • 10 Curhan SG, Willett WC, Grodstein F, Curhan GC. Longitudinal study of hearing loss and subjective cognitive function decline in men. Alzheimers Dement 2019; 15 (04) 525-533
  • 11 Deal JA, Betz J, Yaffe K. et al; Health ABC Study Group. Hearing impairment and incident dementia and cognitive decline in older adults: the Health ABC Study. J Gerontol A Biol Sci Med Sci 2017; 72 (05) 703-709
  • 12 McGilton KS, Höbler F, Campos J. et al. Hearing and vision screening tools for long-term care residents with dementia: protocol for a scoping review. BMJ Open 2016; 6 (07) e011945
  • 13 Cotanche DA. Regeneration of hair cell stereociliary bundles in the chick cochlea following severe acoustic trauma. Hear Res 1987; 30 (2-3): 181-195
  • 14 Cruz RM, Lambert PR, Rubel EW. Light microscopic evidence of hair cell regeneration after gentamicin toxicity in chick cochlea. Arch Otolaryngol Head Neck Surg 1987; 113 (10) 1058-1062
  • 15 Ryals BM, Rubel EW. Hair cell regeneration after acoustic trauma in adult Coturnix quail. Science 1988; 240 (4860): 1774-1776
  • 16 Cox BC, Chai R, Lenoir A. et al. Spontaneous hair cell regeneration in the neonatal mouse cochlea in vivo. Development 2014; 141 (04) 816-829
  • 17 Corwin JT, Cotanche DA. Regeneration of sensory hair cells after acoustic trauma. Science 1988; 240 (4860): 1772-1774
  • 18 Roberson DW, Rubel EW. Cell division in the gerbil cochlea after acoustic trauma. Am J Otol 1994; 15 (01) 28-34
  • 19 Schimmang T, Pirvola U. Coupling the cell cycle to development and regeneration of the inner ear. Semin Cell Dev Biol 2013; 24 (05) 507-513
  • 20 Izumikawa M, Minoda R, Kawamoto K. et al. Auditory hair cell replacement and hearing improvement by Atoh1 gene therapy in deaf mammals. Nat Med 2005; 11 (03) 271-276
  • 21 Yang J, Bouvron S, Lv P, Chi F, Yamoah EN. Functional features of trans-differentiated hair cells mediated by Atoh1 reveals a primordial mechanism. J Neurosci 2012; 32 (11) 3712-3725
  • 22 Liu Z, Dearman JA, Cox BC. et al. Age-dependent in vivo conversion of mouse cochlear pillar and Deiters' cells to immature hair cells by Atoh1 ectopic expression. J Neurosci 2012; 32 (19) 6600-6610
  • 23 Atkinson PJ, Wise AK, Flynn BO, Nayagam BA, Richardson RT. (2014) Hair Cell Regeneration after ATOH1 Gene Therapy in the Cochlea of Profoundly Deaf Adult Guinea Pigs. PLoS ONE 9(7)
  • 24 Bermingham NA, Hassan BA, Price SD. et al. Math1: an essential gene for the generation of inner ear hair cells. Science 1999; 284 (5421): 1837-1841
  • 25 Woods C, Montcouquiol M, Kelley MW. Math1 regulates development of the sensory epithelium in the mammalian cochlea. Nat Neurosci 2004; 7 (12) 1310-1318
  • 26 Zheng JL, Gao WQ. Overexpression of Math1 induces robust production of extra hair cells in postnatal rat inner ears. Nat Neurosci 2000; 3 (06) 580-586
  • 27 Mizutari K, Fujioka M, Hosoya M. et al. Notch inhibition induces cochlear hair cell regeneration and recovery of hearing after acoustic trauma. Neuron 2013; a 77 (01) 58-69
  • 28 Tona Y, Hamaguchi K, Ishikawa M. et al. Therapeutic potential of a gamma-secretase inhibitor for hearing restoration in a guinea pig model with noise-induced hearing loss. BMC Neurosci 2014; 15: 66
  • 29 Salt AN. Pharmacokinetics of drug entry into cochlear fluids. Volta Review 2005; 105 (03) 277-298
  • 30 Maass JC, Gu R, Basch ML. et al. Changes in the regulation of the Notch signaling pathway are temporally correlated with regenerative failure in the mouse cochlea. Front Cell Neurosci 2015; 9: 110
  • 31 Waqas M, Zhang S, He Z, Tang M, Chai R. Role of Wnt and Notch signaling in regulating hair cell regeneration in the cochlea. Front Med 2016; 10 (03) 237-249
  • 32 Shi F, Hu L, Jacques BE, Mulvaney JF, Dabdoub A, Edge AS. β-Catenin is required for hair-cell differentiation in the cochlea. J Neurosci 2014; 34 (19) 6470-6479
  • 33 Shi F, Cheng YF, Wang XL, Edge AS. Beta-catenin up-regulates Atoh1 expression in neural progenitor cells by interaction with an Atoh1 3′ enhancer. J Biol Chem 2010; 285 (01) 392-400
  • 34 Samarajeewa A, Lenz DR, Xie L. et al. Transcriptional response to Wnt activation regulates the regenerative capacity of the mammalian cochlea. Development 2018; 145 (23) 145
  • 35 McLean WJ, McLean DT, Eatock RA, Edge ASB. Distinct capacity for differentiation to inner ear cell types by progenitor cells of the cochlea and vestibular organs. Development 2016; 143 (23) 4381-4393
  • 36 Barker N, van Es JH, Kuipers J. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007; 449 (7165): 1003-1007
  • 37 Sato T, Vries RG, Snippert HJ. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009; 459 (7244): 262-265
  • 38 Yin X, Farin HF, van Es JH, Clevers H, Langer R, Karp JM. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nat Methods 2014; 11 (01) 106-112
  • 39 Chai R, Kuo B, Wang T. et al. Wnt signaling induces proliferation of sensory precursors in the postnatal mouse cochlea. Proc Natl Acad Sci U S A 2012; 109 (21) 8167-8172
  • 40 McLean WJ, Yin X, Lu L. et al. Clonal expansion of Lgr5-positive cells from mammalian cochlea and high-purity generation of sensory hair cells. Cell Rep 2017; 18 (08) 1917-1929
  • 41 Shi F, Kempfle JS, Edge ASB. Wnt-responsive Lgr5-expressing stem cells are hair cell progenitors in the cochlea. J Neurosci 2012; 32 (28) 9639-9648
  • 42 Lee Y-S, Liu F, Segil N. A morphogenetic wave of p27Kip1 transcription directs cell cycle exit during organ of Corti development. Development 2006; 133 (15) 2817-2826
  • 43 Ruben RJ. Development of the inner ear of the mouse: a radioautographic study of terminal mitoses. Acta Otolaryngol 1967; 220: 220 , 1–44
  • 44 Wang Y, Hirose K, Liberman MC. Dynamics of noise-induced cellular injury and repair in the mouse cochlea. J Assoc Res Otolaryngol 2002; 3 (03) 248-268
  • 45 McLean WJ, Hinton AS, Herby JTJ. et al. Improved speech intelligibility in subjects with stable sensorineural hearing loss following intratympanic dosing of FX-322 in a Phase 1b study. Otol Neurotol 2021; 42 (07) e849-e857
  • 46 Lawson GD, Peterson ME. Speech Audiometry. San Diego: CA: Plural Publishing; 2011
  • 47 Thornton AR, Raffin MJM. Speech-discrimination scores modeled as a binomial variable. J Speech Hear Res 1978; 21 (03) 507-518
  • 48 Wilson RH, Abrams HB, Pillion AL. A word-recognition task in multitalker babble using a descending presentation mode from 24 dB to 0 dB signal to babble. J Rehabil Res Dev 2003; 40 (04) 321-327
  • 49 Wilson RH, McArdle R. Intra- and inter-session test, retest reliability of the Words-in-Noise (WIN) test. J Am Acad Audiol 2007; 18 (10) 813-825
  • 50 Salt AN, Gill RM, Plontke SK. Dependence of hearing changes on the dose of intratympanically applied gentamicin: a meta-analysis using mathematical simulations of clinical drug delivery protocols. Laryngoscope 2008; 118 (10) 1793-1800
  • 51 Badri R, Siegel JH, Wright BA. Auditory filter shapes and high-frequency hearing in adults who have impaired speech in noise performance despite clinically normal audiograms. J Acoust Soc Am 2011; 129 (02) 852-863
  • 52 Motlagh Zadeh L, Silbert NH, Sternasty K, Swanepoel W, Hunter LL, Moore DR. Extended high-frequency hearing enhances speech perception in noise. Proc Natl Acad Sci U S A 2019; 116 (47) 23753-23759
  • 53 Katz J. Handbook of Clinical Audiology. 7th ed.. Philadelphia: PA: Lippincott Williams and Wilkins; 2014
  • 54 Souza PE, Boike KT, Witherell K, Tremblay K. Prediction of speech recognition from audibility in older listeners with hearing loss: effects of age, amplification, and background noise. J Am Acad Audiol 2007; 18 (01) 54-65
  • 55 Plontke SK, Biegner T, Kammerer B, Delabar U, Salt AN. Dexamethasone concentration gradients along scala tympani after application to the round window membrane. Otol Neurotol 2008; 29 (03) 401-406
  • 56 Salt AN, Hartsock JJ, Hou J, Piu F. Comparison of the pharmacokinetic properties of triamcinolone and dexamethasone for local therapy of the inner ear. Front Cell Neurosci 2019; 13: 347
  • 57 Salt AN, Plontke SK. Pharmacokinetic principles in the inner ear: Influence of drug properties on intratympanic applications. Hear Res 2018; 368: 28-40
  • 58 Monson B, Buss E. Does extended high-frequency hearing matter in real-world listening?. Hear J 2019; 72: 30-32
  • 59 Monson BB. Benefits of extended high-frequency hearing for speech perception. J Acoust Soc Am 2020; 148: 2540-2540
  • 60 Monson BB, Hunter EJ, Lotto AJ, Story BH. The perceptual significance of high-frequency energy in the human voice. Front Psychol 2014; 5: 587
  • 61 Trine A, Monson BB. Extended High Frequencies Provide Both Spectral and Temporal Information to Improve Speech-in-Speech Recognition. Trends Hear 2020; 24: 2331216520980299