Prevención y diagnóstico precoz de la sordera por ototóxicos

  1. Núñez Batalla, Faustino 1
  2. Jáudenes Casaubón, Carmen 2
  3. Sequí Canet, José Miguel 3
  4. Vivanco Allende, Ana 3
  5. Zubicaray Ugarteche, José 1
  1. 1 Sociedad Española de Otorrinolaringología y Patología Cervico-Facial
    info

    Sociedad Española de Otorrinolaringología y Patología Cervico-Facial

    Madrid, España

  2. 2 Confederación Española de Familias de Personas Sordas (FIAPAS)
  3. 3 Asociación Española de Pediatría
    info

    Asociación Española de Pediatría

    Madrid, España

Revista:
Revista Española de Discapacidad (REDIS)

ISSN: 2340-5104

Año de publicación: 2021

Título del ejemplar: REVISTA ESPAÑOLA DE DISCAPACIDAD | Diciembre 2021 - Mayo 2022

Volumen: 9

Número: 2

Páginas: 155-178

Tipo: Artículo

DOI: 10.5569/2340-5104.09.02.09 DIALNET GOOGLE SCHOLAR lock_openDialnet editor

Otras publicaciones en: Revista Española de Discapacidad (REDIS)

Resumen

Núñez, F. (2021). Prevención y diagnóstico precoz de la sordera por ototóxicos: recomendaciones CODEPEH 2020. Revista Española de Discapacidad, 9(2), pp. 155-178.

Referencias bibliográficas

  • Abujamra, A. L. et al. (2013). The use of high-frequency audiometry increases the diagnosis of asymptomatic hearing loss in pediatric patients treated with cisplatin-based chemotherapy. Pediatr Blood Cancer, 60, pp. 474-478.
  • Arslan, E. et al. (1999). Global problem of drug-induced hearing loss. Ann N Y Acad Sci, 884, pp. 1-14.
  • Astolfi, L. et al. (2016). Coenzyme Q10 plus multivitamin treatment prevents cisplatin ototoxicity in rats. PLoS One, 11(9), p. e0162106.
  • Bass, J. K. y Bhagat, S. P. (2014). Challenges in ototoxicity monitoring in the pediatric oncology population. J Am Acad Audiol, 25, pp. 760-774.
  • Bisht, M. y Bist, S. S. (2011). Ototoxicity: the hidden menace. Indian J Otolaryngol Head Neck Surg, 63, pp. 255-259.
  • Bitner-Glindzicz, M. et al. (2009). Prevalence of mitochondrial 1555A-->G mutation in European children. N Engl J Med, 360(6), pp. 640-642.
  • Brock, P. R. et al. (2012). 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, 30, pp. 2408-2417.
  • Budai, B. et al. (2020). Géczi GSTM1 null and GSTT1 null: Predictors of cisplatin-caused acute ototoxicity measured by DPOAEs. J Mol Med (Berl), 98(7), pp. 963-971.
  • Cianfrone, G. et al. (2011). Pharmacological drugs inducing ototoxicity, vestibular symptoms and tinnitus: A reasoned and updated guide. Eur Rev Med Pharmacol Sci, 15(6), pp. 601-636.
  • Clemens, E. et al. (2019). Recommendations for ototoxicity surveillance for childhood, adolescent, and young adult cancer survivors: A report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCare Consortium. Lancet Oncol, 20, pp. E29-E41.
  • Clemens, E. et al. (2020). Genetic variation of cisplatin-induced ototoxicity in non-cranial-irradiated pediatric patients using a candidate gene approach: The International PanCare LIFE Study. Pharmacogenomics J, 20(2), pp. 294-305.
  • Coradini, P. P. et al. (2007). Ototoxicity from cisplatin therapy in childhood cancer. J Pediatr Hematol Oncol, 29, pp. 355-360.
  • Crabb, S. J. et al. (2017). COAST (Cisplatin ototoxicity attenuated by aspirin trial): A phase II double-blind, randomised controlled trial to establish if aspirin reduces cisplatin induced hearing-loss. Eur J Cancer, 87, pp. 75-83.
  • Crundwell, G. et al. (2016). Ototoxicity (cochleotoxicity) classifications: A review. Int J Audiol, 55(2), pp. 65-74. Davis, B. D. (1987). Mechanism of bactericidal action of aminoglycosides. Microbiol Rev, 51, pp. 341-350.
  • Dogan, M. et al. (2017). Protective role of misoprostol in prevention of gentamicin ototoxicity. Int J Pediatr Otorhinolaryngol, 96, pp. 140-144.
  • Dogan, M. et al. (2018). Utilizing prestin as a predictive marker for the early detection of outer hair cell damage. Am J Otolaryngol, 39(5), pp. 594-598.
  • Dos Santos, N. A. G. et al. (2020). Overview of cisplatin-induced neurotoxicity and ototoxicity, and the protective agents. Food Chem Toxicol, 136, p. 111079.
  • Drögemöller, B. I. et al. (2018). Further Investigation of the Role of ACYP2 and WFS1 Pharmacogenomic Variants in the Development of Cisplatin-Induced Ototoxicity in Testicular Cancer Patients. Clin Cancer Res, 24(8), pp. 1866-1871.
  • Dulon, D. et al. (1988). Relationship between the nephrotoxicity and ototoxicity induced by gentamicin in the Guinea pig. Acta Oto-Laryngologica, 106, pp. 219-225.
  • Dulon, D. et al. (2013). Ototoxicidad farmacológica. EMC-Otorrinolaringología, 42, pp. 1-13.
  • Duval, M. y Daniel, S-J. (2012). Meta-analysis of the efficacy of amifostine in the prevention of cisplatin ototoxicity. Journal of Otolaryngology-Head and Neck Surgery, 41(5), pp. 309-315.
  • El-Anwar, M. W. et al. (2018). Protective effect of pentoxifylline on amikacin-induced ototoxicity. Ear NoseThroat J, 97(8), pp. E8-E12.
  • El-Barbary, M. N. et al. (2015). Gentamicin extended interval regimen and ototoxicity in neonates. Int J Pediatr Otorhinolaryngol, 79, pp. 1294-1298.
  • Fligor, B. J. (2019). Pediatric ototoxicity: current trends and management. Seminars in Hearing, 40, pp. 154-161.
  • Forge, A. y Li, L. (2000). Apoptotic death of hair cells in mammalian vestibular sensory epithelia. Hear Res, 139, pp. 97-115.
  • Fox, D. J. et al. (2016). D-Methionine reduces tobramycin-induced ototoxicity without antimicrobial interference in animal models. Journal of Cystic Fibrosis, 15(4), pp. 518-530.
  • Fransson, A. E. et al. (2017). Hydrogen inhalation protects against ototoxicity induced by intravenous cisplatin in the Guinea pig. Front Cell Neurosci, 11, p. 280.
  • Freyer, D. R. et al. (2017). Effects of sodium thiosulfate versus observation on development of cisplatin-induced hearing loss in children with cancer (ACCL0431): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncology, 18(1), pp. 63-74.
  • Ganesan, P. et al. (2018). Ototoxicity: A challenge in diagnosis and treatment. J Audiol Otol, 22, pp. 59-68.
  • Gallo-Terán, J. et al. (2002). Incidence of A1555G mutations in the mitochondrial DNA and 35delG in the GJB2 gene (connexin-26) in families with late onset non-syndromic sensorineural hearing loss from Cantabria. Acta Otorrinolaringol Esp, 53(8), pp. 563-571.
  • Gu, J. et al. (2020). Astaxanthin-loaded polymer-lipid hybrid nanoparticles (ATX-LPN): Assessment of potential otoprotective effects. J Nanobiotechnology, 18(1), p. 53.
  • Guo, J. et al. (2019a). Protection of hair cells from ototoxic drug-induced hearing loss. En H. Li y R. Chai (Eds.), Hearing loss: Mechanisms, prevention and cure (pp. 17-36). Springer Nature Singapore.
  • Guo, S. et al. (2019b). Rapamycin protects spiral ganglion neurons from gentamicin-induced degeneration in vitro. J Assoc Res Otolaryngol, 20(5), pp. 475-487.
  • Hamstra, D. A. et al. (2010). Pharmacokinetic analysis and phase 1 study of MRX-1024 in patients treated with radiation therapy with or without cisplatinum for head and neck cancer. Clin Cancer Res, 16, pp. 2666-2676.
  • Hana, R. S. y Bawi B. L. (2018). Prestin, otolin-1 regulation, and human 8-oxoG DNA glycosylase 1 gene polymorphisms in noise-induced hearing loss. Ibnosina J Med Biomed Sci, 10, pp. 60-64.
  • Hirvonen, T. P. et al. (2005). Effects of intratympanic gentamicin on vestibular afferents and hair cells in the chinchilla. J Neurophysiol, 93(2), pp. 643-655.
  • Holzer, A. K. et al. (2004). The copper influx transporter human copper transport protein 1 regulates the uptake of cisplatin in human ovarian carcinoma cells. Mol Pharmacol, 66, pp. 817-823.
  • Huang, S. et al. (2013). Proton pump inhibitor selectively suppresses proliferation and restores the chemosensitivity of gastric cancer cells by inhibiting STAT3 signaling pathway. International Immunopharmacology, 17(3), pp. 585-592.
  • Husain, K. et al. (2005). Partial protection by lipoic acid against carboplantin-induced ototoxicity in rats. Biomedical and Environmental Sciences, 18(3), pp. 198-206.
  • Igumnova, V. et al. (2019). The prevalence of mitochondrial mutations associated with aminoglycoside-induced deafness in ethnic Latvian population: The appraisal of the evidence. J Hum Genet, 64(3), pp. 199-206.
  • Ishikawa, M. et al. (2019). Lower ototoxicity and absence of hidden hearing loss point to gentamicin C1a and apramycin as promising antibiotics for clinical use. Sci Rep, 9(1), p. 2410.
  • Jing, W. et al. (2015). Mitochondrial mutations associated with aminoglycoside ototoxicity and hearing loss susceptibility identified by meta-analysis. J Med Genet, 52, pp. 95-103.
  • Juhn, S. K. et al. (1981). Nature of blood-labyrinth barrier in experimental conditions. Ann Otol Rhinol Laryngol, 90, pp. 135-141.
  • Kil, J. E. et al. (2017). Safety and efficacy of ebselen for the prevention of noise-induced hearing loss: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet, 390(10098), pp. 969-979.
  • King, K. A. y Brewer, C. C. (2018). Clinical trials, ototoxicity grading scales, and the audiologist´s role in therapeutic decision making. Int J Audiol, 57, pp. S89-S98.
  • Knight, K. R. et al. (2005). Ototoxicity in children receiving platinum chemotherapy: underestimating a commonly occurring toxicity that may influence academic and social development. J Clin Oncol, 23(34), pp. 8588-8596.
  • Knight, K. R. et al. (2017). Group-wide, prospective study of ototoxicity assessment in children receiving cisplatin chemotherapy (ACCL05C1): A report from the Children’s Oncology Group. J Clin Oncol, 35(4), pp. 440-445.
  • Kotecha, B. y Richardson, G. P. (1994). Ototoxicity in vitro: Effects of neomycin, gentamicin, dihydrostreptomycin, amikacin, spectinomycin, neamine, spermine and poly-L-lysine. Hear Res, 73, pp. 173-184.
  • Kranzer, K. et al. (2015). A systematic review and meta-analysis of the efficacy and safety of N-acetylcysteine in preventing aminoglycoside-induced ototoxicity: Implications for the treatment of multidrug-resistant TB. Thorax, 70(11), pp. 1070-1077.
  • Kros, C. J. y Steyger, P. S. (2019). Aminoglycoside- and cisplatin-induced ototoxicity: Mechanisms and Otoprotective Strategies. Cold Spring Harb Perspect Med, 9, p. a033548.
  • Kucharava, K. et al. (2019). Pasireotide protects mammalian cochlear hair cells from gentamicin ototoxicity by activating the PI3K-Akt pathway. Cell Death Dis, 10(2), p. 110.
  • Lanvers-Kaminsky, C. et al. (2017). Drug-induced ototoxicity: Mechanisms, pharmacogenetics and protective strategies. Clinical Pharmacology & Therapeutics, 101, pp. 491-500.
  • Lanvers-Kaminsky, C. y Ciarimboli, G. (2017). Pharmacogenetics of drug-induced ototoxicity caused by aminoglycosides and cisplatin. Pharmacogenomics, 18, pp. 1683-1695.
  • Laurell, G. (2019). Pharmacological intervention in the field of ototoxicity. HNO, 67(6), pp. 434-439.
  • Le Prell, C. G. (2019). Otoprotectants: From research to clinical application. Semin Hear, 40(2), pp. 162-176.
  • Lee, J. E. et al. (2004). Signaling pathway for apoptosis of vestibular hair cells of mice due to aminoglycosides. Acta Otolaryngol, 124, pp. 69-74.
  • Lee, S. H. et al. (2018). Circulating serum miRNA-205 as a diagnostic biomarker for ototoxicity in mice treated with aminoglycoside antibiotics. Int J Mol Sci, 19(9), p. E2836.
  • Lewis, M. et al. (2009). Ototoxicity in children treated for osteosarcoma. Pediatric Blood Cancer, 52, pp. 387-391.
  • Ma, W. et al. (2015). Ginkgolide B protects against cisplatin-induced ototoxicity: Enhancement of Akt–Nrf2–HO-1 signaling and reduction of NADPH oxidase. Cancer Chemotherapy and Pharmacology, 75, pp. 949-959.
  • Maru, D. y Malky, G. A. (2018). Current practice of ototoxicity management across the United Kingdom (UK). Int J Audiol, 57(sup4), pp. S76-S88.
  • More, S. S. et al. (2010). Role of the copper transporter, CTR1, in platinum-induced ototoxicity. J Neurosci, 30, pp. 9500-9509.
  • Mulry, E. y Parham, K. (2020). Inner ear proteins as potential biomarkers. Otol Neurotol, 41(2), pp. 145-152.
  • Nguyen, T. y Jeyakumar, A. (2019). Genetic susceptibility to aminoglycoside ototoxicity. Int J Pediatr Otorhinolaryngol, 120, pp. 15-19.
  • Núñez, F. et al. (2015). Recomendaciones CODEPEH 2014 (sorderas diferidas y sobrevenidas en la infancia). Revista Española de Discapacidad, 3(1), pp. 163-186.
  • Olivier, T. W. et al. (2019). Cognitive implications of ototoxicity in pediatric patients with embryonal brain tumors. J Clin Oncol, 37, pp. 1566-1575.
  • O’Reilly, M. et al. (2019). Gentamicin affects the bioenergetics of isolated mitochondria and collapses the mitochondrial membrane potential in cochlear sensory hair cells. Front Cell Neurosci, 13, p. 416.
  • Özel, H. E. et al. (2016). Comparison of the protective effects of intratympanic dexamethasone and methylprednisolone against cisplatin-induced ototoxicity. Journal of Laryngology & Otology, 130(3), pp. 225-234.
  • Paciello, F. et al. (2020). The dual role of curcumin and ferulic acid in counteracting chemoresistance and cisplatininduced ototoxicity. Sci Rep, 10(1), p. 1063.
  • Pham, T. N. M. et al. (2020). Protective mechanisms of avocado oil extract against ototoxicity. Nutrients, 12(4), p. 947.
  • Prayuenyong, P. et al. (2020). The efficacy of statins as otoprotective agents: A systematic review. Clin Otolaryngol, 45(1), pp. 21-31.
  • Ramaswamy, B. et al. (2017). Magnetic nanoparticle mediated steroid delivery mitigates cisplatin induced hearing loss. Front Cell Neurosci, 11, p. 268.
  • Rizzi, M. y Hirose, K. (2007). Aminoglycoside ototoxicity. Current opinions in Otolaryngology and Head and Neck Surgery, 15(5), pp. 352-357.
  • Roland, P. S. y Rutka, J. A. (2004). Ototoxicity. BC Decker.
  • Romano, A. et al. (2020). Assessment and management of platinum-related ototoxicity in children treated for cancer. Cancers (Basel), 12(5), p. 1266.
  • Rybak, L. P. (1993). Ototoxicity of loop diuretics. Otolaryngol Clin North Am, 26, pp. 829-844.
  • Salt, A. N. y Plontke, S. K. (2005). Local inner-ear drug delivery and pharmacokinetics. Drug Discov Today, 10, pp. 1299-1306.
  • Sekulic-Jablanovic, M. et al. (2019). Combination of antioxidants and NFAT (nuclear factor of activated T cells) inhibitor protects auditory hair cells from ototoxic insult. J Neurochem, 22, p. e14921.
  • Sha, S. H. et al. (2006). Aspirin to prevent gentamicin-induced hearing loss. N Engl J Med, 354, pp. 1856-1857.
  • Trendowski, M. R. et al. (2019). Genetic and modifiable risk factors contributing to cisplatin-induced toxicities. Clin Cancer Res, 25(4), pp. 1147-1155.
  • Truong, M. T. et al. (2007). Recovery from cisplatin-induced ototoxicity: A case report and review. Int J Pediatr Otorhinolaryngol, 71, pp. 1631-1638.
  • van As, J. W. et al. (2019). Medical interventions for the prevention of platinum-induced hearing loss in children with cancer. Cochrane Database Syst Rev, 5(5), p. CD009219.
  • Villani, V. et al. (2016). Vitamin E neuroprotection against cisplatin ototoxicity: preliminary results from a randomized, placebo-controlled trial. Head Neck, 38(S1), p. E2118.
  • Vital, I. et al. (2015). Distortion-product otoacoustic emissions testing in neonates treated with an aminoglycoside in a neonatal intensive care unit. Ear Nose Throat J, 94, pp. 156-165.
  • Waissbluth, S. (2020). Clinical trials evaluating transtympanic otoprotectants for cisplatin-induced ototoxicity: What do we know so far? Eur Arch Otorhinolaryngol, 277(9), pp. 2413-2422.
  • Watanabe, K. et al. (2003). Expression of caspase-activated deoxyribonuclease (CAD) and caspase 3 (CPP32) in the cochlea of cisplatin (CDDP)-treated Guinea pigs. Auris Nasus Larynx, 30, pp. 219-225.
  • Weiss, A. et al. (2018). Audiological monitoring in Swiss childhood cancer patients. Pediatr Blood Cancer, 65, p. e26877.
  • Yao, L. et al. (2020). Mechanisms and pharmacokinetic/pharmacodynamic profiles underlying the low nephrotoxicity and ototoxicity of etimicin. Acta Pharmacol Sin, 41(6), pp. 866-878.
  • Yorgason, J. G. et al. (2006). Understanding drug ototoxicity: Molecular insights for prevention and clinical management. Expert Opinion on Drug Safety, 5, pp. 383-399.
  • Yu, X. et al. (2018). Paeoniflorin reduces neomycin-induced ototoxicity in hair cells by suppression of reactive oxygen species generation and extracellularly regulated kinase signalization. Toxicol Lett, 285, pp. 9-19.
  • Zada, S. L. et al. (2020). Chemical modifications reduce auditory cell damage induced by aminoglycoside antibiotics. J Am Chem Soc, 12(6), pp. 3077-3087.