Significance
The Ototoxicity Working Group of Pharmaceutical Interventions for Hearing Loss defined ototoxicity as damage to the inner ear, targeting cochlear and vestibular structures as well as sensory function, due to exposure to certain pharmaceuticals, chemicals, and/or ionizing radiation. Ototoxicity typically focuses on the inner ear; however, ototoxins can also affect central auditory pathways and are, thus, considered neurotoxic1,2. Ototoxins can also cause kidney damage and associated renal dysfunction3–5. This review will focus primarily on ototoxic hospitalbased medications received by several hundred thousand infants and children in Western Europe and the Americas each year – aminoglycoside antibiotics and the anti-cancer drug, cisplatin. Other clinically-relevant drugs are listed in Table 1.
Table 1. Major classes of ototoxic drugs (with specific examples).
Adapted from Steyger PS. Mechanisms of Aminoglycoside- and Cisplatin-induced Ototoxicity. American Journal of Audiology. 2021;in press, with permission from Cold Spring Harbor Press.
Class | Examples |
---|---|
Platinum-based therapeutics | Cisplatin, carboplatin, oxaliplatin |
Aminoglycosides | Amikacin, gentamicin, tobramycin |
Peptide antibiotics | Capreomycin, viomycin, chloramphenicol |
Polypeptide antibiotics | Vancomycin (and synergistic with aminoglycosides) |
Macrolides | Erythromycin |
Cyclodextrins (vehicle) | Derivatives of cyclodextrins, e.g., for Neiman-Pick Syndrome Type 1C |
Anti-malarials | Chloroquine, hydrochloroquine |
Anti-rheumatics | Chloroquine, hydrochloroquine |
Loop diuretics | Furosemide, bumetanide (and synergistic with aminoglycosides or cisplatin) |
Non-steroidal anti-inflammatory drugs (NSAIDs) | Acetaminophen (a.k.a. paracetamol) |
Anti-neoplastics | Vincristine |
Aminoglycoside antibiotics, like gentamicin, are clinically-relevant, broad-spectrum medications to treat life-threatening bacterial infections, often of unknown etiology, including neonatal sepsis, mycobacterial infections, and meningitis. Exacerbated respiratory infections in children with cystic fibrosis are typically treated with tobramycin or amikacin, and individuals with tuberculosis with kanamycin, and these are also aminoglycoside antibiotics. Aminoglycoside-induced hearing loss is typically cumulatively dose-dependent6,7, and the incidence in humans is as high as 20–63% of those receiving multi-day dosing7–11. The prevalence of aminoglycoside-induced vestibular deficits is under-reported due to the widespread lack of clinical instrumentation for assessing vestibular function12.
Liver tumors and glioblastomas in the brains of children are effectively diminished by cisplatin or other platinum-based derivatives like carboplatin and oxaliplatin, and the degree of drug-induced hearing loss is typically dose-dependent13–15. There are fewer reports of cisplatin-induced vestibular deficits in children due to the lack of routine clinical assessments12,16, although there are pre-clinical reports of dose-dependent cisplatin-induced vestibular deficits17. Children with Niemann Pick Type 1C syndrome can be treated with derivatives of cyclodextrins, a primary component of drug formulations, that at higher doses also sequester membrane cholesterols18. Treatment with these pharmacological agents in clinical settings can cause ototoxicity. Environmental ototoxins include solvents19–21, and metals (e.g., lead, copper)22–25.
Uncorrected congenital or acquired hearing loss in infants and children can lead to delayed acquisition of listening and spoken language skills compared to age-matched peers with typical hearing, with concomitant delays in achieving academic, linguistic, and psychosocial milestones26,27. Loss of vestibular function is also debilitating28,29. There is an estimated socioeconomic cost >$1.5 million over the lifetime of each pre-lingually deafened child (in 2019 dollars)30.
Crossing the blood-labyrinth barrier
The inner ear is protected by a blood-labyrinth barrier (BLB; akin to the blood-brain barrier) that compartmentalizes inner ear cells and fluids from the bloodstream and the rest of the body. Endothelial cells lining cochlear blood vessels are coupled together by tight junctions to prevent extravasation (i.e., the paracellular trafficking of blood cells, macromolecules, and serum from the capillary into cochlear tissues)31. Within the cochlea, there are two major fluid spaces, perilymph and endolymph. Blood-borne ototoxins are readily trafficked into the perilymph surrounding the basolateral membranes of hair cells32,33, yet this does not appear to be the primary entry route into hair cells34 (see Figure 1). Instead, aminoglycosides and cisplatin cross the BLB of the stria vascularis and, from there, appear to be rapidly cleared into endolymph and enter hair cells across their apical membranes more readily than when directly infused into perilymph34–37. The cellular and molecular mechanisms by which aminoglycosides and cisplatin cross the BLB and transverse the tight-junction-coupled marginal cells of the stria vascularis into endolymph remain to be determined, and likely include one or more of the following molecular mechanisms: ion channels, transporters, exchangers, or transcytosis12,38, each permutation specific for each individual class of ototoxins.
Figure 1: Primary trafficking routes of aminoglycosides and cisplatin from the vasculature to cochlear hair cells.
Circulating ototoxins typically enter the cochlea via capillaries in the stria vascularis and are cleared into endolymph via as-yet-unidentified ion channels or transporters, although several candidates exist, e.g., TRPV1 and TRPV4 non-selective cation channels for aminoglycosides, and potentially via OCT2 and CTR1 transporters for cisplatin. Once in endolymph, ototoxins can enter hair cells via one or more of several mechanisms.
From Kros CJ, Steyger PS. Aminoglycoside- and Cisplatin-Induced Ototoxicity: Mechanisms and Otoprotective Strategies. Cold Spring Harb Perspect Med. 2019;9(11), with permission from Cold Spring Harbor Press.
Entry of ototoxins into sensory hair cells
Typically, cochleotoxicity requires ototoxins to enter hair cells39. At the distal tips of most stereocilia in the mechanically-sensitive hair bundle are transduction channels (Figure 2A) that allow the polycationic aminoglycosides to permeate into the cell40. These transduction channels are non-selective cation channels such as TMC141. Aminoglycoside permeation of TMC1 channels can be blocked by polyvalent cations like magnesium or calcium (Figure 2B), as well as by organic compounds like curare and quinine42,43. Hair cells also express other aminoglycoside-permeant channels, including several transient receptor potential (TRP) channels, each activated by different physical or chemical stimuli44. These include TRPV1 and TRPV4 at the apical membrane of hair cells (Figure 2C)45–48. The aminoglycoside-permeant TRPA1 channel is likely expressed on the basolateral membranes of outer hair cells (Figure 2D)49. Aminoglycosides can also enter hair cells via non-receptor-mediated endocytosis and are trafficked to lysosomes (Figure 2E). Blocking endocytosis (Figure 2F) or impeding intracellular trafficking of aminoglycoside-laden endosomes does not prevent hair cell death42,50,51. Cisplatin can enter cells via passive diffusion across the cell membrane (Figure 3A)52. Hair cells with active (open) transduction channels (Figure 3B) are more susceptible to cisplatin-induced cytotoxicity53.
Figure 2: Aminoglycoside entry into hair cells.
Aminoglycosides preferentially enter mammalian hair cells via the TMC1 channel that consists of two TMC subunits (purple), each with a permeation groove (A). Entry of aminoglycosides can be blocked by curare, quinine and high levels of polyvalent cations (B). Other aminoglycoside-permeant channels on the apical membrane of hair cells include TRPV1 and TPRV4 (C), and TRPA1 on the basolateral membrane of outer hair cells (D). (E) Non-specific endocytosis enables aminoglycoside-laden endosomes to traffic to hair cell lysosomes. (F) Blocking endocytosis does not prevent hair cell death when aminoglycosides can enter hair cells via the TMC1 channel.
From Steyger PS. Mechanisms of Aminoglycoside- and Cisplatin-induced Ototoxicity. American Journal of Audiology. 2021;in press, with permission from the American Journal of Audiology.
Figure 3: Cisplatin entry into hair cells.
Cisplatin has multiple potential entry routes. (A) Neutral cisplatin can diffuse across the plasma membrane, and is readily aquated in the cytoplasm to the more toxic form of cisplatin that can form functionally disruptive adducts with proteins and DNA. (B) Uptake of aquated cisplatin is dependent on functional TMC channel complexes. (C) Cellular uptake of the aquated form of cisplatin can also occur via CTR1 and OCT2 transport proteins when expressed by the cell.
From Steyger PS. Mechanisms of Aminoglycoside- and Cisplatin-induced Ototoxicity. American Journal of Audiology. 2021;in press, with permission from the American Journal of Audiology.
Once in cochlear hair cells, cochleotoxic dosing with aminoglycosides and cisplatin first leads to outer hair cell death in the basal high frequency region of the cochlea, particularly in outer hair cells. Increasing cumulative dosing leads to further outer hair cell death in more apical regions of the cochlea (i.e., at lower frequencies) and inner hair cell death, with an increasing risk of permanent hearing loss7,15,54. There is one major difference between aminoglycosides and cisplatin, and that is the preferential cochleotoxicity of cisplatin, compared to the equal reporting of cochleotoxicity and vestibulotoxicity induced by aminoglycosides, although the underlying etiology for this difference remains hypothetical55.
There are multiple (yet incompletely-defined) molecular mechanisms of hair cell death, and the best characterizations for aminoglycosides and cisplatin are reviewed elsewhere4,5,12. It is important to stress that ototoxicity is not dependent on hair cell dysfunction or cell death, and can dysregulate non-sensory cells within the inner ear essential for sensitive auditory perception (e.g., the stria vascularis)12. Below, we focus on clinical settings that can exacerbate the degree of hearing loss and vestibular disorders induced by ototoxins, particularly by aminoglycosides and cisplatin.
Clinical settings that can exacerbate the risk of drug-induced hearing loss
Experimentally-induced inflammation (to mimic host-mounted responses to bacterial infection) exacerbates the degree of aminoglycoside- and cisplatin-induced hearing loss in mice38,48,56, and potentially for vestibulotoxicity57. Pilot studies of neonates treated with aminoglycosides likely confirm these preclinical findings58,59. Aminoglycoside-induced lysis of bacteria elevates the inflammatory response (i.e., the Jarisch-Herxheimer reaction)60,61. The mechanisms underlying the potentiation of hearing loss by inflammation remain to be established62,63. Thus, the very patients with bacterial infections (and therefore inflammation) treated with aminoglycosides are likely at higher risk of ototoxin-induced hearing loss38.
Other clinical settings can also exacerbate the risk of drug-induced hearing loss, and include: renal insufficiency decreasing the clearance of ototoxins from blood64,65, increasing age with concommitant decreased glomerular filtration rate66–69; depletion of endogenous antioxidants needed to ameliorate ototoxin-induced generation of toxic levels of reactive oxygen species70–72; fever (hyperthermia or higher-than normal body temperature)59,73, and transient ischemia/hypoxia74. Aminoglycoside- and cisplatin-induced cochleotoxicity is also greater when these ototoxins are administered during active hours (i.e., nighttime) for rodents75–78.
Several single nucleotide polymorphisms in mitochondrial ribosomal RNA (e.g., A1555G and C1494T) result in a higher binding affinity to aminoglycosides, leading to mistranslation of mRNA and inaccurate protein synthesis and a greater susceptibility to aminoglycoside-induced hearing loss79–82. At this time, no genomic polymorphisms have been reported to modulate susceptibility to aminoglycoside-induced hearing loss. Several genomic polymorphisms have been reported to increase the risk of cisplatin-induced hearing loss, including gene variants for antioxidant enzymes, DNA adduct repair enzymes; antioxidant enzymes, drug efflux or membrane pumps83–86. However, the predictive value of these genomic variants in predisposing individuals for drug-induced ototoxicity is currently poor87,88.
Hospitalized children frequently receive multiple medications simultaneously, and several clinically-relevant non-ototoxic drugs can potentiate drug-induced hearing loss. For example, the neuromuscular blocking agent pancuronium bromide is frequently prescribed for neonates requiring respiratory assistance (via intubation and ventilation); however, pancuronium bromide can potentiate the cochleotoxicity of loop diuretics prescribed for hypervolemic patients89,90. Simultaneous dosing with two or more ototoxic therapeutics can synergistically exacerbate the degree of ototoxicity to greater than the sum of the two ototoxins alone. These co-therapeutics include aminoglycosides and loop diuretics in hypertensive individuals91–93, or aminoglycosides with vancomycin, a glycopeptide antibiotic commonly prescribed in the neonatal intensive care unit94,95. Enhanced ototoxicity can also occur when cisplatin is co-administered with aminoglycosides, loop diuretics, and cranial radiation96–99. Thus, when dosing with multiple drugs simultaneously, all therapeutics should be examined for toxic interactions, including potentiated or synergistic ototoxic effects, and alternative dosing regimens identified where possible.
Loud sound exposures exacerbate the degree of aminoglycoside-induced ototoxicity100–102. Noise exposure also increases cisplatin-induced hearing loss103–106. These data are relevant when children require pharmacotherapy following recent exposure to intense loud sounds.
Otoprotection
Strategies that protect the inner ear from the toxic effects of ototoxins in children are currently considered to be the same as being developed for adults. Multiple strategies have been investigated, although few have completed Phase III clinical trials and none are yet approved by the Federal Drug Administration (FDA) in the United States of America. The most-intensely investigated strategy to prevent ototoxicity has been efforts to inhibit the toxic generation of reactive oxygen/nitrogen species or the production of proapoptotic factors15,107,108. Potentially more specific, otoprotective strategies now include blocking systemically-administered ototoxins from cross the BLB and entering the inner ear fluids to prevent toxicity12, and physicochemical structural modification of ototoxins to prevent entry into hair cells and thus ameliorate ototoxicity while retaining their desired efficacy have also been investigated109,110, although for derivatives of cisplatin, this has led to reduced clinical efficacy (i.e., toxicity to the tumor) despite decreased ototoxicity15.
Alternative delivery strategies have focused on local delivery to the middle or inner ear to reduce the systemic toxicity associated with oral or parenteral administration of candidate otoprotective compounds ; however, local delivery has challenges too, principally rapid clearance from the middle or inner ear111. One strategy to reduce clearance has been to use hydrogels and nanoparticles to retain the drug in the vicinity of the round window membrane. One novel strategy is to inject drug-loaded magnetic nanoparticles onto the round window and use a contralateral magnet to pull the magnetic nanoparticles into the inner ear. This strategy abrogated cisplatin-induced hearing loss in the high frequency (basal) region of the cochlea112.
Unbiased screening
Many of the above strategies to prevent ototoxicity used a rational, deductive approach to selecting and designing otoprotective dosing regimens, and have been clearly successful preclinically to varying degrees. An alternative strategy has emerged in recent years to use unbiased screening of large compound libraries in silico, in vitro and/or using zebrafish larvae to identify promising ototoxic “hits” or candidate otoprotective compounds.
Computational, or in silico, models use molecular structures and physiochemical properties to predict drug toxicity or efficacy in an unbiased manner, based on reported structure-activity relationships. This approach has been used to predict cardio- or hepatotoxicity113,114. Two studies have reported in silico screening strategies for ototoxicity, while others use pathway or linkage models (https://lincsproject.org/LINCS) to identify potential otoprotectants62,115. Promising hits or candidate otoprotective compounds identified in silico must be verified using in vitro or in vivo assays. In vitro assays typically use cell lines, including those derived from the cochlea116–118. Although cell lines do not possess stereocilia with mechanically-gated TMC1 channels, cell lines are extremely useful for deciphering the molecular and signaling pathways underlying ototoxic or otoprotective mechanisms118–121.
Like most fish, zebrafish possess on the surface of their body a mechanosensitive lateral line consisting of neuromasts within which are sensory hair cells with stereocilia and mechanically-gated transduction channels, analogous to cochlear and vestibular hair cells. Female zebrafish release hundreds of larvae in each clutch, and within 4–5 days after fertilization have functional neuromasts122. These numbers, and ease of access (without a BLB) enable medium-throughput screening of compound libraries to identify hits that induce hair cell toxicity62, or interact with hair cell transduction channels to protect neuromast hair cells from aminoglycoside or cisplatin cytotoxicity123–125. Subsequently, these hits or candidate otoprotective compounds need to be validated preclinically using inner ear explants and in vivo ototoxicity preclinical models, primarily in rodents and higher- order mammals, before advancing to human clinical trials117,118,124.
Challenges to preventing ototoxicity
We do not yet appreciate the numerous ways each ototoxin can induce cytotoxicity. This is best exemplified by the hundreds of observed aminoglycoside- or cisplatin-binding proteins, most of which remain unidentified and therefore uncharacterized, of which a fraction will have cytotoxic interactions and the remainder incidental, benign, or even cytoprotective interactions120,121,126. Hair cells and strial cells (like renal proximal tubule cells) are often unable to clear ototoxins from their cytosol35,127. Most other cells within the body are typically able to clear ototoxins from their cytosol, often by as-yet-unknown mechanisms; the reasons for these differences in toxicologic sensitivities remain highly speculative.
If candidate otoprotective compounds are to be delivered orally or systemically, and the primary site of otoprotection is within the inner ear itself, these compounds must first cross the BLB at sufficient quantities to meet therapeutic levels without being systemically toxic. If this is not possible, local delivery via intratympanic administration might be viable if the candidate otoprotective compounds are efficacious in perilymph. Other crucial research-driven questions have been posed elsewhere102.
Although knowledge of the mechanism by which therapeutic compounds achieve their efficacy is not needed for FDA approval, the mechanisms by which candidate compounds provide their otoprotective effect are increasingly being requested. This can add years to the discovery pipeline before a drug can be advanced to clinical trials. This phenomenon can be abrogated by repurposing already-FDA-approved drugs for other indications to off-label indications if otoprotection can be shown preclinically (e.g. statins)128,129. Candidate otoprotective compounds must be effective in the medical settings in which they will be used (e.g., in individuals with host-mounted inflammatory responses to infection or cancer treatment (e.g. radiation). This practice has been robustly implemented for pharmaceutical interventions for loud sound exposures, but not for prevention of ototoxicity in sick individuals.
Several non-pharmacological, yet otoprotective or rehabilitative interventions, are also likely in the near future. The identification of risk factors, including aging, renal insufficiency, inflammation, genetic polymorphisms, or selected co-therapeutics, that predispose individuals to a greater risk of ototoxicity can be extracted from the medical record or tested for. For example, clinical guidelines exist for identifying which currently known mitochondrial polymorphisms predispose individuals to ototoxicity82. In such cases, non-ototoxic pharmacotherapies can be prescribed. Even if these alternate routes can be more expensive in the short-term, they would still be much cheaper than the socioeconomic costs over the individual’s lifetime. Additional non-pharmacological otoprotective strategies include developing point-of-care (bedside) diagnostic procedures for sepsis through species-specific detection of microbial DNA sequences130 or genomic screening for genomic polymorphisms that enhance susceptibility to drug-induced ototoxicity86,131.
Nonetheless, at present, the prevalence and incidence of most predisposing factors remain uncertain due to the variability of each subject’s medical and ototoxin- dosing history within a population. Given the need to prescribe ototoxic medications to individuals in life-threatening situations, it is important to implement ototoxicity monitoring protocols as recommended for those with cystic fibrosis132. Widespread implementation of ototoxicity monitoring protocols will provide new insights into the prevalence and incidence of cochleotoxicity and vestibulotoxicity that will accelerate the relative significance and impact of ototoxicity on quality of life scores and optimal rehabilitation of those affected by drug-induced hearing loss.
Key Points:
Numerous hospital-prescribed medications and environmental factors cause ototoxicity.
Ototoxicity encompasses hearing loss (cochleotoxicity) and/or balance deficits (vestibulotoxicity).
Ototoxicity has permanent, life-long debilitating consequences, and if uncorrected can lead to unfulfilled scholastic and career trajectories in children, as well as accelerated cognitive decline in aging individuals.
Otoprotection to preserve or restore auditory and/or vestibular function includes ototoxicity monitoring, prosthetic and social rehabilitation, and soon, pharmaceutical ototherapeutics to preserve, repair, or restore inner ear functions.
Ototoxicity is, ideally, a preventable form of acquired hearing loss or vestibular deficit.
Synopsis.
Ototoxicity refers to damage to the inner ear that leads to functional hearing loss or vestibular disorders by selected pharmacotherapeutics as well as a variety of environmental exposures (e.g., lead, cadmium, solvents). This chapter reviews fundamental mechanisms underlying ototoxicity by clinically-relevant, hospital-prescribed medications (i.e., aminoglycoside antibiotics or cisplatin, as illustrative examples). Also reviewed are current strategies to prevent prescribed medication-induced ototoxicity, with several clinical or candidate interventional strategies being discussed.
Clinics Care Points.
The degree of expected ototoxicity from multi- day dosing with known ototoxins can be amplified by risk factors such as renal insufficiency or systemic inflammation.
Dosing with multiple pharmacotherapeutics simultaneously (even if separated temporally) can exacerbate the degree of ototoxicity of a known ototoxin.
Ototoxicity monitoring is vital when dosing with life-saving ototoxins, especially as any identified hearing loss can then be more rapidly rehabilitated, ameliorating the negative impact of drug-induced hearing loss.
Disclosure Statement:
The author has no conflicts of interests pertinent to this manuscript. This review was supported by NIH-NIDCD research awards R01 DC004555 and R01 DC016880, as well as an NIGMS CoBRE Award P20 GM139762 to Peter S. Steyger, PhD.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Gopal KV, Wu C, Shrestha B, Campbell KC, Moore EJ, Gross GW. d-Methionine protects against cisplatin-induced neurotoxicity in cortical networks. Neurotoxicol Teratol. 2012;34(5):495–504. [DOI] [PubMed] [Google Scholar]
- 2.Hinduja S, Kraus KS, Manohar S, Salvi RJ. D-methionine protects against cisplatin-induced neurotoxicity in the hippocampus of the adult rat. Neurotoxicity research. 2015;27(3):199–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Humes HD. Insights into ototoxicity. Analogies to nephrotoxicity. Ann N Y Acad Sci. 1999;884:1518. [DOI] [PubMed] [Google Scholar]
- 4.Jiang M, Karasawa T, Steyger PS. Aminoglycoside-Induced Cochleotoxicity: A Review. Frontiers in cellular neuroscience. 2017;11:308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Karasawa T, Steyger PS. An integrated view of cisplatin-induced nephrotoxicity and ototoxicity. Toxicol Lett. 2015;237(3):219–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wu WJ, Sha SH, McLaren JD, Kawamoto K, Raphael Y, Schacht J. Aminoglycoside ototoxicity in adult CBA, C57BL and BALB mice and the Sprague-Dawley rat. Hear Res. 2001;158(1–2):165–178. [DOI] [PubMed] [Google Scholar]
- 7.Garinis AC, Cross CP, Srikanth P, et al. The cumulative effects of intravenous antibiotic treatments on hearing in patients with cystic fibrosis. Journal of cystic fibrosis : official journal of the European Cystic Fibrosis Society. 2017;16(3):401–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Garinis AC, Liao S, Cross CP, et al. Effect of gentamicin and levels of ambient sound on hearing screening outcomes in the neonatal intensive care unit: A pilot study. Int J Pediatr Otorhinolaryngol. 2017;97:42–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fausti SA, Frey RH, Henry JA, Olson DJ, Schaffer HI. Early detection of ototoxicity using highfrequency, tone-burst-evoked auditory brainstem responses. J Am Acad Audiol. 1992;3(6):397404. [PubMed] [Google Scholar]
- 10.Fausti SA, Henry JA, Schaffer HI, Olson DJ, Frey RH, McDonald WJ. High-frequency audiometric monitoring for early detection of aminoglycoside ototoxicity. J Infect Dis. 1992;165(6):10261032. [DOI] [PubMed] [Google Scholar]
- 11.Elson EC, Meier E, Oermann CM. The implementation of an aminoglycoside induced ototoxicity algorithm for people with cystic fibrosis. Journal of cystic fibrosis : official journal of the European Cystic Fibrosis Society. 2021, 20 (2) 284–287. [DOI] [PubMed] [Google Scholar]
- 12.Kros CJ, Steyger PS. Aminoglycoside- and Cisplatin-Induced Ototoxicity: Mechanisms and Otoprotective Strategies. Cold Spring Harb Perspect Med. 2019;9(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Knight KR, Kraemer DF, Neuwelt EA. Ototoxicity in children receiving platinum chemotherapy: underestimating a commonly occurring toxicity that may influence academic and social development. J Clin Oncol. 2005;23(34):8588–8596. [DOI] [PubMed] [Google Scholar]
- 14.Knight KR, Kraemer DF, Winter C, Neuwelt EA. Early changes in auditory function as a result of platinum chemotherapy: use of extended high-frequency audiometry and evoked distortion product otoacoustic emissions. J Clin Oncol. 2007;25(10):1190–1195. [DOI] [PubMed] [Google Scholar]
- 15.Brock PR, Knight KR, Freyer DR, et al. Platinum-induced ototoxicity in children: a consensus review on mechanisms, predisposition, and protection, including a new International Society of Pediatric Oncology Boston ototoxicity scale. J Clin Oncol. 2012;30(19):2408–2417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Prayuenyong P, Taylor JA, Pearson SE, et al. Vestibulotoxicity Associated With Platinum-Based Chemotherapy in Survivors of Cancer: A Scoping Review. Frontiers in Oncology. 2018;8:363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Callejo A, Durochat A, Bressieux S, et al. Dose-dependent cochlear and vestibular toxicity of trans-tympanic cisplatin in the rat. Neurotoxicology. 2017;60:1–9. [DOI] [PubMed] [Google Scholar]
- 18.Crumling MA, King KA, Duncan RK. Cyclodextrins and Iatrogenic Hearing Loss: New Drugs with Significant Risk. Frontiers in cellular neuroscience. 2017;11:355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Davis RR, Murphy WJ, Snawder JE, et al. Susceptibility to the ototoxic properties of toluene is species specific. Hear Res. 2002;166(1–2):24–32. [DOI] [PubMed] [Google Scholar]
- 20.Crofton KM, Lassiter TL, Rebert CS. Solvent-induced ototoxicity in rats: an atypical selective mid- frequency hearing deficit. Hear Res. 1994;80(1):25–30. [DOI] [PubMed] [Google Scholar]
- 21.Gagnaire F, Langlais C. Relative ototoxicity of 21 aromatic solvents. Arch Toxicol. 2005;79(6):346354. [DOI] [PubMed] [Google Scholar]
- 22.Counter SA, Buchanan LH. Neuro-ototoxicity in andean adults with chronic lead and noise exposure. Journal of occupational and environmental medicine / American College of Occupational and Environmental Medicine. 2002;44(1):30–38. [DOI] [PubMed] [Google Scholar]
- 23.Jamesdaniel S, Rosati R, Westrick J, Ruden DM. Chronic lead exposure induces cochlear oxidative stress and potentiates noise-induced hearing loss. Toxicol Lett. 2018;292:175–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hernandez PP, Moreno V, Olivari FA, Allende ML. Sub-lethal concentrations of waterborne copper are toxic to lateral line neuromasts in zebrafish (Danio rerio). Hear Res. 2006;213(1–2):110. [DOI] [PubMed] [Google Scholar]
- 25.Olivari FA, Hernandez PP, Allende ML. Acute copper exposure induces oxidative stress and cell death in lateral line hair cells of zebrafish larvae. Brain Res. 2008;1244:1–12. [DOI] [PubMed] [Google Scholar]
- 26.Dedhia K, Kitsko D, Sabo D, Chi DH. Children with sensorineural hearing loss after passing the newborn hearing screen. JAMA Otolaryngol Head Neck Surg. 2013;139(2):119–123. [DOI] [PubMed] [Google Scholar]
- 27.Gurney JG, Tersak JM, Ness KK, et al. Hearing loss, quality of life, and academic problems in long-term neuroblastoma survivors: a report from the Children’s Oncology Group. Pediatrics. 2007;120(5):e1229–1236. [DOI] [PubMed] [Google Scholar]
- 28.Smith PF, Zheng Y. From ear to uncertainty: vestibular contributions to cognitive function. Front Integr Neurosci. 2013;7:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Smith PF, Darlington CL. Personality changes in patients with vestibular dysfunction. Front Hum Neurosci. 2013;7:678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mohr PE, Feldman JJ, Dunbar JL, et al. The societal costs of severe to profound hearing loss in the United States. Int J Technol Assess Health Care. 2000;16(4):1120–1135. [DOI] [PubMed] [Google Scholar]
- 31.Nyberg S, Abbott NJ, Shi X, Steyger PS, Dabdoub A. Delivery of therapeutics to the inner ear: The challenge of the blood-labyrinth barrier. Science translational medicine. 2019;11(482). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tran Ba Huy P, Bernard P, Schacht J. Kinetics of gentamicin uptake and release in the rat. Comparison of inner ear tissues and fluids with other organs. J Clin Invest. 1986;77(5):14921500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hellberg V, Wallin I, Ehrsson H, Laurell G. Cochlear pharmacokinetics of cisplatin: an in vivo study in the guinea pig. Laryngoscope. 2013;123(12):3172–3177. [DOI] [PubMed] [Google Scholar]
- 34.Li H, Steyger PS. Systemic aminoglycosides are trafficked via endolymph into cochlear hair cells. Sci Rep. 2011;1:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Breglio AM, Rusheen AE, Shide ED, et al. Cisplatin is retained in the cochlea indefinitely following chemotherapy. Nature communications. 2017;8(1):1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chu YH, Sibrian-Vazquez M, Escobedo JO, et al. Systemic Delivery and Biodistribution of Cisplatin in Vivo. Mol Pharm. 2016;13(8):2677–2682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.van Ruijven MW, de Groot JC, Hendriksen F, Smoorenburg GF. Immunohistochemical detection of platinated DNA in the cochlea of cisplatin-treated guinea pigs. Hear Res. 2005;203(1–2):112121. [DOI] [PubMed] [Google Scholar]
- 38.Koo JW, Quintanilla-Dieck L, Jiang M, et al. Endotoxemia-mediated inflammation potentiates aminoglycoside-induced ototoxicity. Science translational medicine. 2015;7(298):298ra118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hiel H, Erre JP, Aurousseau C, Bouali R, Dulon D, Aran JM. Gentamicin uptake by cochlear hair cells precedes hearing impairment during chronic treatment. Audiology. 1993;32(1):78–87. [DOI] [PubMed] [Google Scholar]
- 40.Marcotti W, van Netten SM, Kros CJ. The aminoglycoside antibiotic dihydrostreptomycin rapidly enters mouse outer hair cells through the mechano-electrical transducer channels. J Physiol. 2005;567(Pt 2):505–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pan B, Akyuz N, Liu XP, et al. TMC1 Forms the Pore of Mechanosensory Transduction Channels in Vertebrate Inner Ear Hair Cells. Neuron. 2018;99(4):736–753 e736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Alharazneh A, Luk L, Huth M, et al. Functional hair cell mechanotransducer channels are required for aminoglycoside ototoxicity. PLoS ONE. 2011;6(7):e22347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Coffin AB, Reinhart KE, Owens KN, Raible DW, Rubel EW. Extracellular divalent cations modulate aminoglycoside-induced hair cell death in the zebrafish lateral line. Hear Res. 2009;253(1–2):4251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev. 2014;66(3):676–814. [DOI] [PubMed] [Google Scholar]
- 45.Karasawa T, Wang Q, Fu Y, Cohen DM, Steyger PS. TRPV4 enhances the cellular uptake of aminoglycoside antibiotics. J Cell Sci. 2008;121(Pt 17):2871–2879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Myrdal SE, Steyger PS. TRPV1 regulators mediate gentamicin penetration of cultured kidney cells. Hear Res. 2005;204(1–2):170–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zheng J, Dai C, Steyger PS, et al. Vanilloid receptors in hearing: altered cochlear sensitivity by vanilloids and expression of TRPV1 in the organ of corti. J Neurophysiol. 2003;90(1):444–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jiang M, Li H, Johnson A, et al. Inflammation up-regulates cochlear expression of TRPV1 to potentiate drug-induced hearing loss. Sci Adv. 2019;5(7):eaaw1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Stepanyan RS, Indzhykulian AA, Velez-Ortega AC, et al. TRPA1-mediated accumulation of aminoglycosides in mouse cochlear outer hair cells. J Assoc Res Otolaryngol. 2011;12(6):729740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Vu AA, Nadaraja GS, Huth ME, et al. Integrity and regeneration of mechanotransduction machinery regulate aminoglycoside entry and sensory cell death. PLoS ONE. 2013;8(1):e54794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hailey DW, Esterberg R, Linbo TH, Rubel EW, Raible DW. Fluorescent aminoglycosides reveal intracellular trafficking routes in mechanosensory hair cells. J Clin Invest. 2017;127(2):472–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hall MD, Okabe M, Shen DW, Liang XJ, Gottesman MM. The role of cellular accumulation in determining sensitivity to platinum-based chemotherapy. Annual review of pharmacology and toxicology. 2008;48:495–535. [DOI] [PubMed] [Google Scholar]
- 53.Thomas AJ, Hailey DW, Stawicki TM, et al. Functional mechanotransduction is required for cisplatin-induced hair cell death in the zebrafish lateral line. J Neurosci. 2013;33(10):4405–4414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Forge A, Schacht J. Aminoglycoside antibiotics. Audiol Neurootol. 2000;5(1):3–22. [DOI] [PubMed] [Google Scholar]
- 55.Prayuenyong Pattarawadee, Baguley David, and K C, Steyger PS. Preferential cochleotoxicity of cisplatin. Frontiers in neuroscience. 2021. July 26;15:695268. doi: 10.3389/fnins.2021.695268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Oh GS, Kim HJ, Choi JH, et al. Activation of lipopolysaccharide-TLR4 signaling accelerates the ototoxic potential of cisplatin in mice. J Immunol. 2011;186(2):1140–1150. [DOI] [PubMed] [Google Scholar]
- 57.Qian X, He Z, Wang Y, et al. Hair cell uptake of gentamicin in the developing mouse utricle. J Cell Physiol. 2021;236(7):5235–5252. [DOI] [PubMed] [Google Scholar]
- 58.Cross CP, Liao S, Urdang ZD, Srikanth P, Garinis AC, Steyger PS. Effect of sepsis and systemic inflammatory response syndrome on neonatal hearing screening outcomes following gentamicin exposure. Int J Pediatr Otorhinolaryngol. 2015;79(11):1915–1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Henry KR, Guess MB, Chole RA. Hyperthermia increases aminoglycoside ototoxicity. Acta Otolaryngol. 1983;95(3–4):323–327. [DOI] [PubMed] [Google Scholar]
- 60.Kaplanski G, Granel B, Vaz T, Durand JM. Jarisch-Herxheimer reaction complicating the treatment of chronic Q fever endocarditis: elevated TNFalpha and IL-6 serum levels. The Journal of infection. 1998;37(1):83–84. [DOI] [PubMed] [Google Scholar]
- 61.Shenep JL, Mogan KA. Kinetics of endotoxin release during antibiotic therapy for experimental gram-negative bacterial sepsis. J Infect Dis. 1984;150(3):380–388. [DOI] [PubMed] [Google Scholar]
- 62.Coffin AB, Boney R, Hill JD, Tian C, Steyger PS. Detecting novel ototoxins and potentiation of ototoxicity by disease settings. Front. Neurol, 17 August 2021. | 10.3389/fneur.2021.725566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Jiang M, Taghizadeh F, Steyger PS. Potential Mechanisms Underlying Inflammation-Enhanced Aminoglycoside-Induced Cochleotoxicity. Frontiers in cellular neuroscience. 2017;11:362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zager RA. Endotoxemia, renal hypoperfusion, and fever: interactive risk factors for aminoglycoside and sepsis-associated acute renal failure. Am J Kidney Dis. 1992;20(3):223–230. [DOI] [PubMed] [Google Scholar]
- 65.Gandara DR, Perez EA, Weibe V, De Gregorio MW. Cisplatin chemoprotection and rescue: pharmacologic modulation of toxicity. Seminars in oncology. 1991;18(1 Suppl 3):49–55. [PubMed] [Google Scholar]
- 66.Gatell JM, Ferran F, Araujo V, et al. Univariate and multivariate analyses of risk factors predisposing to auditory toxicity in patients receiving aminoglycosides. Antimicrob Agents Chemother. 1987;31(9):1383–1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Manian FA, Stone WJ, Alford RH. Adverse antibiotic effects associated with renal insufficiency. Reviews of infectious diseases. 1990;12(2):236–249. [DOI] [PubMed] [Google Scholar]
- 68.McClure CL. Common infections in the elderly. Am Fam Physician. 1992;45(6):2691–2698. [PubMed] [Google Scholar]
- 69.Triggs E, Charles B. Pharmacokinetics and therapeutic drug monitoring of gentamicin in the elderly. Clin Pharmacokinet. 1999;37(4):331–341. [DOI] [PubMed] [Google Scholar]
- 70.Lautermann J, McLaren J, Schacht J. Glutathione protection against gentamicin ototoxicity depends on nutritional status. Hear Res. 1995;86(1–2):15–24. [DOI] [PubMed] [Google Scholar]
- 71.Lautermann J, Crann SA, McLaren J, Schacht J. Glutathione-dependent antioxidant systems in the mammalian inner ear: effects of aging, ototoxic drugs and noise. Hear Res. 1997;114(12):75–82. [DOI] [PubMed] [Google Scholar]
- 72.Lautermann J, Song B, McLaren J, Schacht J. Diet is a risk factor in cisplatin ototoxicity. Hear Res. 1995;88(1–2):47–53. [DOI] [PubMed] [Google Scholar]
- 73.Spankovich C, Lobarinas E, Ding D, Salvi R, Le Prell CG. Assessment of thermal treatment via irrigation of external ear to reduce cisplatin-induced hearing loss. Hear Res. 2016;332:55–60. [DOI] [PubMed] [Google Scholar]
- 74.Lin CD, Kao MC, Tsai MH, et al. Transient ischemia/hypoxia enhances gentamicin ototoxicity via caspase-dependent cell death pathway. Lab Invest. 2011;91(7):1092–1106. [DOI] [PubMed] [Google Scholar]
- 75.McKinney W, Yonovitz A, Smolensky MH. Circadian variation of gentamicin toxicity in rats. Laryngoscope. 2015;125(7):E252–256. [DOI] [PubMed] [Google Scholar]
- 76.Soulban G, Smolensky MH, Yonovitz A. Gentamicin-induced chronotoxicity: use of body temperature as a circadian marker rhythm. Chronobiol Int. 1990;7(5–6):393–402. [DOI] [PubMed] [Google Scholar]
- 77.Bielefeld EC, Markle A, DeBacker JR, Harrison RT. Chronotolerance for cisplatin ototoxicity in the rat. Hear Res. 2018;370:16–21. [DOI] [PubMed] [Google Scholar]
- 78.Tserga E, Moreno-Paublete R, Sarlus H, et al. Circadian vulnerability of cisplatin-induced ototoxicity in the cochlea. FASEB J. 2020;34(10):13978–13992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hobbie SN, Bruell CM, Akshay S, Kalapala SK, Shcherbakov D, Bottger EC. Mitochondrial deafness alleles confer misreading of the genetic code. Proc Natl Acad Sci U S A. 2008;105(9):3244–3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Qian Y, Guan MX. Interaction of aminoglycosides with human mitochondrial 12S rRNA carrying the deafness-associated mutation. Antimicrob Agents Chemother. 2009;53(11):4612–4618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Matt T, Ng CL, Lang K, et al. Dissociation of antibacterial activity and aminoglycoside ototoxicity in the 4-monosubstituted 2-deoxystreptamine apramycin. Proc Natl Acad Sci U S A. 2012;109(27):10984–10989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.McDermott JH, Wolf J, Hoshitsuki K, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for the use of aminoglycosides based on MT-RNR1 genotype. J Neuroinflammation. 2021. January 6;18(1):16. doi: 10.1186/s12974-020-02051-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Turan C, Kantar M, Aktan C, et al. Cisplatin ototoxicity in children: risk factors and its relationship with polymorphisms of DNA repair genes ERCC1, ERCC2, and XRCC1. Cancer Chemother Pharmacol. 2019;84(6):1333–1338. [DOI] [PubMed] [Google Scholar]
- 84.Caronia D, Patino-Garcia A, Milne RL, et al. Common variations in ERCC2 are associated with response to cisplatin chemotherapy and clinical outcome in osteosarcoma patients. The pharmacogenomics journal. 2009;9(5):347–353. [DOI] [PubMed] [Google Scholar]
- 85.Xu H, Robinson GW, Huang J, et al. Common variants in ACYP2 influence susceptibility to cisplatin-induced hearing loss. Nat Genet. 2015;47(3):263–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ross CJ, Katzov-Eckert H, Dube MP, et al. Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy. Nat Genet. 2009;41(12):13451349. [DOI] [PubMed] [Google Scholar]
- 87.Langer T, Clemens E, Broer L, et al. Usefulness of current candidate genetic markers to identify childhood cancer patients at risk for platinum-induced ototoxicity: Results of the European PanCareLIFE cohort study. Eur J Cancer. 2020;138:212–224. [DOI] [PubMed] [Google Scholar]
- 88.Tserga E, Nandwani T, Edvall NK, et al. The genetic vulnerability to cisplatin ototoxicity: a systematic review. Sci Rep. 2019;9(1):3455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Masumoto K, Nagata K, Uesugi T, Yamada T, Taguchi T. Risk factors for sensorineural hearing loss in survivors with severe congenital diaphragmatic hernia. European journal of pediatrics. 2007;166(6):607–612. [DOI] [PubMed] [Google Scholar]
- 90.Cheung PY, Tyebkhan JM, Peliowski A, Ainsworth W, Robertson CM. Prolonged use of pancuronium bromide and sensorineural hearing loss in childhood survivors of congenital diaphragmatic hernia. J Pediatr. 1999;135(2 Pt 1):233–239. [DOI] [PubMed] [Google Scholar]
- 91.Mathog RH, Klein WJ Jr., Ototoxicity of ethacrynic acid and aminoglycoside antibiotics in uremia. N Engl J Med. 1969;280(22):1223–1224. [DOI] [PubMed] [Google Scholar]
- 92.Rybak LP. Ototoxicity of loop diuretics. Otolaryngol Clin North Am. 1993;26(5):829–844. [PubMed] [Google Scholar]
- 93.Bates DE, Beaumont SJ, Baylis BW. Ototoxicity induced by gentamicin and furosemide. The Annals of pharmacotherapy. 2002;36(3):446–451. [DOI] [PubMed] [Google Scholar]
- 94.Rubin LG, Sanchez PJ, Siegel J, et al. Evaluation and treatment of neonates with suspected lateonset sepsis: a survey of neonatologists’ practices. Pediatrics. 2002;110(4):e42. [DOI] [PubMed] [Google Scholar]
- 95.Brummett RE, Fox KE, Jacobs F, Kempton JB, Stokes Z, Richmond AB. Augmented gentamicin ototoxicity induced by vancomycin in guinea pigs. Arch Otolaryngol Head Neck Surg. 1990;116(1):61–64. [DOI] [PubMed] [Google Scholar]
- 96.Clemens E, de Vries AC, Pluijm SF, et al. Determinants of ototoxicity in 451 platinum-treated Dutch survivors of childhood cancer: A DCOG late-effects study. Eur J Cancer. 2016;69:77–85. [DOI] [PubMed] [Google Scholar]
- 97.McAlpine D, Johnstone BM. The ototoxic mechanism of cisplatin. Hear Res. 1990;47(3):191–203. [DOI] [PubMed] [Google Scholar]
- 98.Miller MW, Riedel G, Hoistad D, et al. Ototoxicity after combined platinum and fractionated radiation in a novel guinea pig model. Am J Otolaryngol. 2009;30(1):1–7. [DOI] [PubMed] [Google Scholar]
- 99.Paulino AC, Lobo M, Teh BS, et al. Ototoxicity after intensity-modulated radiation therapy and cisplatin-based chemotherapy in children with medulloblastoma. International journal of radiation oncology, biology, physics. 2010;78(5):1445–1450. [DOI] [PubMed] [Google Scholar]
- 100.Collins PW. Synergistic interactions of gentamicin and pure tones causing cochlear hair cell loss in pigmented guinea pigs. Hear Res. 1988;36(2–3):249–259. [DOI] [PubMed] [Google Scholar]
- 101.Li H, Steyger PS. Synergistic ototoxicity due to noise exposure and aminoglycoside antibiotics. Noise Health. 2009;11(42):26–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Steyger PS. Mechanisms of Aminoglycoside- and Cisplatin-induced Ototoxicity. Am J Audiol. 2021. August 20:1–14. doi: 10.1044/2021_AJA-21-00006. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gratton MA, Salvi RJ, Kamen BA, Saunders SS. Interaction of cisplatin and noise on the peripheral auditory system. Hear Res. 1990;50(1–2):211–223. [DOI] [PubMed] [Google Scholar]
- 104.Bokemeyer C, Berger CC, Hartmann JT, et al. Analysis of risk factors for cisplatin-induced ototoxicity in patients with testicular cancer. Br J Cancer. 1998;77(8):1355–1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Laurell GF. Combined effects of noise and cisplatin: short- and long-term follow-up. Ann Otol Rhinol Laryngol. 1992;101(12):969–976. [DOI] [PubMed] [Google Scholar]
- 106.DeBacker JR, Harrison RT, Bielefeld EC. Long-Term Synergistic Interaction of Cisplatin- and NoiseInduced Hearing Losses. Ear Hear. 2017;38(3):282–291. [DOI] [PubMed] [Google Scholar]
- 107.Huth ME, Ricci AJ, Cheng AG. Mechanisms of aminoglycoside ototoxicity and targets of hair cell protection. International journal of otolaryngology. 2011;2011:937861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Mechanisms of Cisplatin-Induced Ototoxicity and Otoprotection. Frontiers in cellular neuroscience. 2017;11:338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Huth ME, Han KH, Sotoudeh K, et al. Designer aminoglycosides prevent cochlear hair cell loss and hearing loss. J Clin Invest. 2015;125(2):583–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.O’Sullivan ME, Song Y, Greenhouse R, et al. Dissociating antibacterial from ototoxic effects of gentamicin C-subtypes. Proc Natl Acad Sci U S A. 2020;117(51):32423–32432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Salt AN, Plontke SK. Principles of local drug delivery to the inner ear. Audiol Neurootol. 2009;14(6):350–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ramaswamy B, Roy S, Apolo AB, Shapiro B, Depireux DA. Magnetic Nanoparticle Mediated Steroid Delivery Mitigates Cisplatin Induced Hearing Loss. Frontiers in cellular neuroscience. 2017;11:268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hammann F, Schoning V, Drewe J. Prediction of clinically relevant drug-induced liver injury from structure using machine learning. J Appl Toxicol. 2019;39(3):412–419. [DOI] [PubMed] [Google Scholar]
- 114.Cai C, Guo P, Zhou Y, et al. Deep Learning-Based Prediction of Drug-Induced Cardiotoxicity. J Chem Inf Model. 2019;59(3):1073–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ryals M, Morell RJ, Martin D, et al. The Inner Ear Heat Shock Transcriptional Signature Identifies Compounds That Protect Against Aminoglycoside Ototoxicity. Frontiers in cellular neuroscience. 2018;12:445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kalinec GM, Webster P, Lim DJ, Kalinec F. A cochlear cell line as an in vitro system for drug ototoxicity screening. Audiol Neurootol. 2003;8(4):177–189. [DOI] [PubMed] [Google Scholar]
- 117.Ingersoll MA, Malloy EA, Caster LE, et al. BRAF inhibition protects against hearing loss in mice. Sci Adv. 2020;6(49). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Teitz T, Fang J, Goktug AN, et al. CDK2 inhibitors as candidate therapeutics for cisplatin- and noise-induced hearing loss. J Exp Med. 2018;215(4):1187–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Hazlitt RA, Teitz T, Bonga JD, et al. Development of Second-Generation CDK2 Inhibitors for the Prevention of Cisplatin-Induced Hearing Loss. J Med Chem. 2018. September 13; 61(17): 7700–7709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Karasawa T, Wang Q, David LL, Steyger PS. CLIMP-63 is a gentamicin-binding protein that is involved in drug-induced cytotoxicity. Cell Death Dis. 2010;1:e102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Karasawa T, Wang Q, David LL, Steyger PS. Calreticulin binds to gentamicin and reduces drug-induced ototoxicity. Toxicol Sci. 2011;124(2):378–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Harris JA, Cheng AG, Cunningham LL, MacDonald G, Raible DW, Rubel EW. Neomycin-induced hair cell death and rapid regeneration in the lateral line of zebrafish (Danio rerio). J Assoc Res Otolaryngol. 2003;4(2):219–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Owens KN, Santos F, Roberts B, et al. Identification of genetic and chemical modulators of zebrafish mechanosensory hair cell death. PLoS genetics. 2008;4(2):e1000020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Chowdhury S, Owens KN, Herr RJ, et al. Phenotypic Optimization of Urea-Thiophene Carboxamides To Yield Potent, Well Tolerated, and Orally Active Protective Agents against Aminoglycoside-Induced Hearing Loss. J Med Chem. 2018;61(1):84–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kenyon EJ, Kirkwood NK, Kitcher SR, et al. Identification of ion-channel modulators that protect against aminoglycoside-induced hair cell death. JCI Insight. 2017;2(24). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Karasawa T, Sibrian-Vazquez M, Strongin RM, Steyger PS. Identification of cisplatin-binding proteins using agarose conjugates of platinum compounds. PLoS ONE. 2013;8(6):e66220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Dai CF, Mangiardi D, Cotanche DA, Steyger PS. Uptake of fluorescent gentamicin by vertebrate sensory cells in vivo. Hear Res. 2006;213(1–2):64–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Fernandez K, Spielbauer KK, Rusheen A, et al. Lovastatin protects against cisplatin-induced hearing loss in mice. Hear Res. 2020;389:107905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Fernandez KA, Allen P, Campbell M, et al. Atorvastatin is associated with reduced cisplatin-induced hearing loss. J Clin Invest. 2021;131(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Trung NT, Hien TT, Huyen TT, et al. Enrichment of bacterial DNA for the diagnosis of blood stream infections. BMC infectious diseases. 2016;16:235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Jing W, Zongjie H, Denggang F, et al. Mitochondrial mutations associated with aminoglycoside ototoxicity and hearing loss susceptibility identified by meta-analysis. J Med Genet. 2015;52(2):95–103. [DOI] [PubMed] [Google Scholar]
- 132.Garinis AC, Poling GL, Rubenstein RC, et al. Clinical Considerations for Routine Auditory and Vestibular Monitoring in Patients with Cystic Fibrosis. American Journal of Audiology. 2021;in press. [DOI] [PMC free article] [PubMed] [Google Scholar]