1
|
Ehlert K, Heinze B, Graham MA, Swanepoel DW. Changes in vestibular and cochlear function following platinum-based chemotherapy: A preliminary report. EAR, NOSE & THROAT JOURNAL 2025; 104:NP337-NP348. [PMID: 36052943 DOI: 10.1177/01455613221115042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
ObjectivesThis study investigated the changes in vestibular and cochlear function in patients receiving platinum-based chemotherapy.MethodsA longitudinal study of 32 participants (10-70 years) receiving chemotherapy participated in the study. Baseline and exit vestibular and hearing assessments that included video head impulse (VHIT) testing, cervical and ocular vestibular evoked myogenic potentials (VEMP), dynamic visual acuity (DVA) and pure-tone audiometry were performed at the patient's treatment venue.ResultsHalf (50%) of the participants showed cochleotoxicity from baseline to exit testing, with left ears significantly more affected than right ears. There was no consistent relationship between hearing loss and vestibular dysfunction. DVA yielded normal results at baseline and exit testing in all participants. VEMP responses were absent in 28.1% of participants at baseline, reflecting the challenges of using VEMP for monitoring. VEMP and VHIT results showed a statistically significant (p < 0.05) decline in results from baseline to exit testing; however, participants did not report symptoms related to vestibular dysfunction. VHIT also showed left ears significantly (p < 0.05) more affected than right ears.ConclusionVHIT proved to be a valuable measure of changes in vestibular function secondary to ototoxicity. Future investigations should determine vestibulotoxicity criteria and optimal protocols for vestibulotoxicity monitoring at the patient's treatment venue.
Collapse
Affiliation(s)
- Katerina Ehlert
- Department Speech-Language Pathology and Audiology, University of Pretoria, Pretoria, South Africa
- Department Speech-Language Pathology and Audiology, Sefako Makgatho Health Sciences University, South Africa
| | - Barbara Heinze
- Department Speech-Language Pathology and Audiology, University of Pretoria, Pretoria, South Africa
- Ear Science Institute Australia, Subiaco, WA, Australia
| | - Marien A Graham
- Department of Science, Mathematics and Technology Education, University of Pretoria, Pretoria, South Africa
| | - De Wet Swanepoel
- Department Speech-Language Pathology and Audiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
2
|
Salah RS, Mahmoud AA, El-Shiekh RA, El-Dessouki AM, Hassan AGAE, Khalaf SS. A comprehensive review of the impact of natural products in preventing drug-induced ototoxicity. Inflammopharmacology 2025:10.1007/s10787-025-01766-2. [PMID: 40338449 DOI: 10.1007/s10787-025-01766-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025]
Abstract
Ototoxicity, the property of certain drugs to cause hearing loss, is a significant concern in medical treatments, particularly with the use of chemotherapeutic agents like cisplatin and aminoglycosides. These drugs can lead to permanent sensorineural hearing loss (SNHL), affecting a substantial proportion of patients. Existing strategies to alleviate these side effects are limited, prompting interest in natural products as potential protective agents. Natural products are being investigated for their ability to counteract these mechanisms through anti-inflammatory and antioxidant properties. The review seeks to highlight the potential of these natural products as complementary therapies to conventional ototoxic medications, emphasizing their protective roles, which are involved in cochlear cellular damage and programmed cell death. Further research is essential to establish standardized protocols for their use and to ensure their integration into clinical practice as effective therapeutic options.
Collapse
Affiliation(s)
- Rania S Salah
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Asmaa A Mahmoud
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy, Al-Azhar University (Girls), Cairo, 11884, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt
| | | | - Samar S Khalaf
- Biochemistry Department Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
3
|
Kallenberger EM, Briggs EE, Nguyen SA, Dixon PR, Drawdy AV, Labadie RF, Meyer TA, White DR. Preventing Hearing Loss in Children Receiving Cisplatin: A Systematic Review and Meta-Analysis. Laryngoscope 2025. [PMID: 40165641 DOI: 10.1002/lary.32158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/05/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE Cisplatin-induced hearing loss leads to significant neurologic, social, and behavioral impairment in children. The goal of this study is to characterize options available to prevent cisplatin-induced hearing loss and to identify gaps in the literature. DATA SOURCES CINAHL, Cochrane Library, PubMed, Scopus. REVIEW METHODS Literature was searched between 1990 and 2024. Studies evaluating interventions to prevent hearing loss in children receiving cisplatin were included. Audiometric data including pure tone threshold, pure tone average, and incidence of hearing loss were extracted from included studies. RESULTS Six studies (N = 760) pertaining to cisplatin-induced hearing loss in children were included. This includes four randomized control trials (N = 652), one nonrandomized control trial (N = 97) and one prospective cohort study (N = 11). The studies examined sodium thiosulfate or amifostine, both given intravenously. The relative risk of hearing loss between intervention and control groups was 0.78 (95% CI 0.71-0.85). The proportion of patients in the treatment group categorized as grade zero on the Brock ototoxicity grading scale was significantly higher (p < 0.0001) than in the control group (36.3% vs. 15.5%). The change in pure tone average after chemotherapy was significantly higher in the control group compared to the intervention group (5.2 vs. -1.2 dB, 95% CI 5.53-7.25). CONCLUSIONS These results show mild success in reducing the incidence of hearing loss in children undergoing chemotherapy with cisplatin. However, the literature is limited, and further investigation is warranted.
Collapse
Affiliation(s)
- Ethan M Kallenberger
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Erin E Briggs
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
- Sydney Kimmel Medical College, Thomas Jefferson University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shaun A Nguyen
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Peter R Dixon
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Allyson V Drawdy
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina School of Medicine, St. Charleston, South Carolina, USA
| | - Robert F Labadie
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ted A Meyer
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David R White
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
4
|
Manavi MA, Fathian Nasab MH, Mohammad Jafari R, Dehpour AR. Mechanisms underlying dose-limiting toxicities of conventional chemotherapeutic agents. J Chemother 2024; 36:623-653. [PMID: 38179685 DOI: 10.1080/1120009x.2023.2300217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Dose-limiting toxicities (DLTs) are severe adverse effects that define the maximum tolerated dose of a cancer drug. In addition to the specific mechanisms of each drug, common contributing factors include inflammation, apoptosis, ion imbalances, and tissue-specific enzyme deficiencies. Among various DLTs are bleomycin-induced pulmonary fibrosis, doxorubicin-induced cardiomyopathy, cisplatin-induced nephrotoxicity, methotrexate-induced hepatotoxicity, vincristine-induced neurotoxicity, paclitaxel-induced peripheral neuropathy, and irinotecan, which elicits severe diarrhea. Currently, specific treatments beyond dose reduction are lacking for most toxicities. Further research on cellular and molecular pathways is imperative to improve their management. This review synthesizes preclinical and clinical data on the pharmacological mechanisms underlying DLTs and explores possible treatment approaches. A comprehensive perspective reveals knowledge gaps and emphasizes the need for future studies to develop more targeted strategies for mitigating these dose-dependent adverse effects. This could allow the safer administration of fully efficacious doses to maximize patient survival.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
6
|
Fleihan T, Nader ME, Dickman JD. Cisplatin vestibulotoxicity: a current review. Front Surg 2024; 11:1437468. [PMID: 39421409 PMCID: PMC11484025 DOI: 10.3389/fsurg.2024.1437468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Cisplatin, a commonly used chemotherapy drug, is well-established for its ototoxic effects, primarily attributed to the damage it inflicts on cochlear hair cells. However, its impact on the vestibular system remains inadequately understood. Here, we provide a comprehensive review of existing literature concerning cisplatin-induced vestibulotoxicity. Animal studies have shown that cisplatin induces a vestibular hair cell loss that is dose-dependent, with the severity of damage also varying according to the route of administration. Notably, intratympanic and systemic injections in animal models have manifested significant damage primarily to utricular hair cells, with a lesser degree of damage observed for the other vestibular end organs. The underlying mechanisms of cisplatin induced vestibular hair cell loss include apoptosis, oxidative stress, and inflammatory cytokines. Several protective agents, such as Pifithrin-α, DAPT, Ginkgolide B, and heat shock proteins, have demonstrated efficacy in inhibiting cisplatin-induced vestibular damage in preclinical studies. Human clinical findings indicate that cisplatin treatment can cause vestibular dysfunction, characterized by symptoms ranging from transient dizziness to persistent vertigo. Challenges in diagnosis, including the limited utilization of comprehensive vestibular testing for many patients, contribute to the variability in reported outcomes. Cisplatin-induced vestibulotoxicity is a significant complication of chemotherapy, necessitating further research to understand its mechanisms and to improve diagnosis and management, ultimately aiming to enhance the quality of life for cancer patients undergoing cisplatin therapy.
Collapse
Affiliation(s)
- Tamara Fleihan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Marc Elie Nader
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - J. David Dickman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
7
|
Kessler L, Koo C, Richter CP, Tan X. Hearing loss during chemotherapy: prevalence, mechanisms, and protection. Am J Cancer Res 2024; 14:4597-4632. [PMID: 39417180 PMCID: PMC11477841 DOI: 10.62347/okgq4382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Ototoxicity is an often-underestimated sequela for cancer patients undergoing chemotherapy, with an incidence rate exceeding 50%, affecting approximately 4 million individuals worldwide each year. Despite the nearly 2,000 publications on chemotherapy-related ototoxicity in the past decade, the understanding of its prevalence, mechanisms, and preventative or therapeutic measures remains ambiguous and subject to debate. To date, only one drug, sodium thiosulfate, has gained FDA approval for treating ototoxicity in chemotherapy. However, its utilization is restricted. This review aims to offer clinicians and researchers a comprehensive perspective by thoroughly and carefully reviewing available data and current evidence. Chemotherapy-induced ototoxicity is characterized by four primary symptoms: hearing loss, tinnitus, vertigo, and dizziness, originating from both auditory and vestibular systems. Hearing loss is the predominant symptom. Amongst over 700 chemotherapeutic agents documented in various databases, only seven are reported to induce hearing loss. While the molecular mechanisms of the hearing loss caused by the two platinum-based drugs are extensively explored, the pathways behind the action of the other five drugs are primarily speculative, rooted in their therapeutic properties and side effects. Cisplatin attracts the majority of attention among these drugs, encompassing around two-thirds of the literature regarding ototoxicity in chemotherapy. Cisplatin ototoxicity chiefly manifests through the loss of outer hair cells, possibly resulting from damages directly by cisplatin uptake or secondary effects on the stria vascularis. Both direct and indirect influences contribute to cisplatin ototoxicity, while it is still debated which path is dominant or where the primary target of cisplatin is located. Candidates for hearing protection against cisplatin ototoxicity are also discussed, with novel strategies and methods showing promise on the horizon.
Collapse
Affiliation(s)
- Lexie Kessler
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Chail Koo
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Claus-Peter Richter
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Biomedical Engineering, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Communication Sciences and Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| | - Xiaodong Tan
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| |
Collapse
|
8
|
Shankar V, Seethapathy J, Srinivas S, Nandhan R, Saravanam PK. Prevailing Practices in Ototoxicity Monitoring in Individuals With Head and Neck Cancer Undergoing Radiotherapy and Chemoradiotherapy: A Scoping Review. Am J Audiol 2024; 33:1041-1069. [PMID: 39052439 DOI: 10.1044/2024_aja-24-00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
PURPOSE The aim of the current scoping review is to identify the studies reporting ototoxicity monitoring in individuals with head and neck cancer (HNC) undergoing radiation therapy and/or chemoradiation therapy across the world. The specific objectives were to identify and report the test protocol used, identify the most common timeline of follow-up, and identify barriers and facilitators influencing the implementation of the monitoring program. METHOD A comprehensive search was conducted across six electronic databases, including PubMed, Embase, Web of Science, Scopus, Google Scholar, and ProQuest. The scoping review method adhered to relevant guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews) and frameworks. The database search was carried out by two independent researchers, and studies were selected based on specific inclusion and exclusion criteria. RESULTS This scoping review identified 13 studies that fulfilled the inclusion criteria of this study. Only one study reported findings from the perspective of ototoxicity monitoring. Another study explicitly mentioned that ototoxicity monitoring was a standard of care in their hospital. Only one study reported using the relevant guidelines for monitoring ototoxicity. Specialized tests such as high-frequency audiometry, distortion product otoacoustic emissions, and vestibular tests were rarely used. Ototoxicity monitoring was influenced by awareness-related factors, technical factors, treatment-related factors, and organizational factors. CONCLUSIONS Research on ototoxicity monitoring programs is in its early stages, highlighting the need for standardized practices and multidisciplinary collaboration to enhance health care services for HNC patients. A standardized approach, improved awareness, and the incorporation of patient perspectives are crucial to enhancing ototoxicity monitoring in HNC patients.
Collapse
Affiliation(s)
- Varsha Shankar
- Department of Audiology, Sri Ramachandra Faculty of Audiology and Speech Language Pathology, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, India
| | - Jayashree Seethapathy
- Department of Audiology, Sri Ramachandra Faculty of Audiology and Speech Language Pathology, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, India
| | - Satish Srinivas
- Department of Radiation Oncology, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, India
| | - Raghu Nandhan
- Department of ENT-Head & Neck Surgery, Madras ENT Research Foundation, Chennai, Tamil Nadu, India
| | - Prasanna Kumar Saravanam
- Department of ENT, Head and Neck Surgery, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, India
| |
Collapse
|
9
|
Sung CYW, Hayase N, Yuen PST, Lee J, Fernandez K, Hu X, Cheng H, Star RA, Warchol ME, Cunningham LL. Macrophage depletion protects against cisplatin-induced ototoxicity and nephrotoxicity. SCIENCE ADVANCES 2024; 10:eadk9878. [PMID: 39047106 PMCID: PMC11268410 DOI: 10.1126/sciadv.adk9878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Cisplatin is a widely used anticancer drug with notable side effects including ototoxicity and nephrotoxicity. Macrophages, the major resident immune cells in the cochlea and kidney, are important drivers of both inflammatory and tissue repair responses. To investigate the roles of macrophages in cisplatin-induced toxicities, we used PLX3397, a U.S. Food and Drug Administration-approved inhibitor of the colony-stimulating factor 1 receptor, to eliminate tissue-resident macrophages. Mice treated with cisplatin alone had considerable hearing loss (ototoxicity) and kidney injury (nephrotoxicity). Macrophage ablation resulted in significantly reduced hearing loss and had greater outer hair cell survival. Macrophage ablation also protected against cisplatin-induced nephrotoxicity, as evidenced by markedly reduced tubular injury and fibrosis. Mechanistically, our data suggest that the protective effect of macrophage ablation against cisplatin-induced ototoxicity and nephrotoxicity is mediated by reduced platinum accumulation in both the inner ear and the kidney. Together, our data indicate that ablation of tissue-resident macrophages represents an important strategy for mitigating cisplatin-induced ototoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- Cathy Yea Won Sung
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Naoki Hayase
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Peter S. T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - John Lee
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Katharine Fernandez
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Hui Cheng
- Bioinformatics and Biostatistics Collaboration Core, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Mark E. Warchol
- Department of Otolaryngology, School of Medicine, Washington University, Saint Louis, MO, USA
| | - Lisa L. Cunningham
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| |
Collapse
|
10
|
Sanchez VA, Dinh PC, Monahan PO, Althouse S, Rooker J, Sesso HD, Dolan ME, Weinzerl M, Feldman DR, Fung C, Einhorn LH, Frisina RD, Travis LB. Comprehensive Audiologic Analyses After Cisplatin-Based Chemotherapy. JAMA Oncol 2024; 10:912-922. [PMID: 38842797 PMCID: PMC11157440 DOI: 10.1001/jamaoncol.2024.1233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/22/2023] [Indexed: 06/07/2024]
Abstract
Importance Cisplatin is highly ototoxic but widely used. Evidence is lacking regarding cisplatin-related hearing loss (CRHL) in adult-onset cancer survivors with comprehensive audiologic assessments (eg, Words-in-Noise [WIN] tests, full-spectrum audiometry, and additional otologic measures), as well as the progression of CRHL considering comorbidities, modifiable factors associated with risk, and cumulative cisplatin dose. Objective To assess CRHL with comprehensive audiologic assessments, including the WIN, evaluate the longitudinal progression of CRHL, and identify factors associated with risk. Design, Setting, and Participants The Platinum Study is a longitudinal study of cisplatin-treated testicular cancer survivors (TCS) enrolled from 2012 to 2018 with follow-up ongoing. Longitudinal comprehensive audiologic assessments at Indiana University and Memorial Sloan Kettering Cancer Center included 100 participants without audiometrically defined profound hearing loss (HL) at baseline and at least 3.5 years from their first audiologic assessment. Data were analyzed from December 2013 to December 2022. Exposures Factors associated with risk included cumulative cisplatin dose, hypertension, hypercholesterolemia, diabetes, tobacco use, physical inactivity, body mass index, family history of HL, cognitive dysfunction, psychosocial symptoms, and tinnitus. Main Outcomes and Measures Main outcomes were audiometrically measured HL defined as combined-ears high-frequency pure-tone average (4-12 kHz) and speech-recognition in noise performance measured with WIN. Multivariable analyses evaluated factors associated with risk for WIN scores and progression of audiometrically defined HL. Results Median (range) age of 100 participants at evaluation was 48 (25-67) years; median (range) time since chemotherapy: 14 (4-31) years. At follow-up, 78 (78%) TCS had audiometrically defined HL; those self-reporting HL had 2-fold worse hearing than TCS without self-reported HL (48 vs 24 dB HL; P < .001). A total of 54 (54%) patients with self-reported HL showed clinically significant functional impairment on WIN testing. Poorer WIN performance was associated with hypercholesterolemia (β = 0.88; 95% CI, 0.08 to 1.69; P = .03), lower-education (F1 = 5.95; P = .004), and severity of audiometrically defined HL (β̂ = 0.07; 95% CI, 0.06 to 0.09; P < .001). CRHL progression was associated with hypercholesterolemia (β̂ = -4.38; 95% CI, -7.42 to -1.34; P = .01) and increasing age (β̂ = 0.33; 95% CI, 0.15 to 0.50; P < .001). Importantly, relative to age-matched male normative data, audiometrically defined CRHL progression significantly interacted with cumulative cisplatin dose (F1 = 5.98; P = .02); patients given 300 mg/m2 or less experienced significantly less progression, whereas greater temporal progression followed doses greater than 300 mg/m2. Conclusions and Relevance Follow-up of cisplatin-treated cancer survivors should include strict hypercholesterolemia control and regular audiological assessments. Risk stratification through validated instruments should include querying hearing concerns. CRHL progression relative to age-matched norms is likely associated with cumulative cisplatin dose; investigation over longer follow-up is warranted.
Collapse
Affiliation(s)
- Victoria A. Sanchez
- Department of Otolaryngology–Head and Neck Surgery, University of South Florida, Tampa
| | - Paul C. Dinh
- Department of Medical Oncology, Indiana University, Indianapolis
| | - Patrick O. Monahan
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis
| | - Sandra Althouse
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis
| | | | - Howard D. Sesso
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - M. Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Mandy Weinzerl
- Rehabilitation Services, Indiana University Health, Indianapolis
| | - Darren R. Feldman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chunkit Fung
- Department of Medical Oncology, J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | - Robert D. Frisina
- Department of Medical Engineering, University of South Florida, Tampa
| | - Lois B. Travis
- Department of Medical Oncology, Indiana University, Indianapolis
| |
Collapse
|
11
|
Knecht DA, Zeziulia M, Bhavsar MB, Puchkov D, Maier H, Jentsch TJ. LRRC8/VRAC volume-regulated anion channels are crucial for hearing. J Biol Chem 2024; 300:107436. [PMID: 38838775 PMCID: PMC11260850 DOI: 10.1016/j.jbc.2024.107436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Hearing crucially depends on cochlear ion homeostasis as evident from deafness elicited by mutations in various genes encoding cation or anion channels and transporters. Ablation of ClC‑K/barttin chloride channels causes deafness by interfering with the positive electrical potential of the endolymph, but roles of other anion channels in the inner ear have not been studied. Here we report the intracochlear distribution of all five LRRC8 subunits of VRAC, a volume-regulated anion channel that transports chloride, metabolites, and drugs such as the ototoxic anti-cancer drug cisplatin, and explore its physiological role by ablating its subunits. Sensory hair cells express all LRRC8 isoforms, whereas only LRRC8A, D and E were found in the potassium-secreting epithelium of the stria vascularis. Cochlear disruption of the essential LRRC8A subunit, or combined ablation of LRRC8D and E, resulted in cochlear degeneration and congenital deafness of Lrrc8a-/- mice. It was associated with a progressive degeneration of the organ of Corti and its innervating spiral ganglion. Like disruption of ClC-K/barttin, loss of VRAC severely reduced the endocochlear potential. However, the mechanism underlying this reduction seems different. Disruption of VRAC, but not ClC-K/barttin, led to an almost complete loss of Kir4.1 (KCNJ10), a strial K+ channel crucial for the generation of the endocochlear potential. The strong downregulation of Kir4.1 might be secondary to a loss of VRAC-mediated transport of metabolites regulating inner ear redox potential such as glutathione. Our study extends the knowledge of the role of cochlear ion transport in hearing and ototoxicity.
Collapse
Affiliation(s)
- Deborah A Knecht
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Mariia Zeziulia
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; Graduate Program of the Freie Universität Berlin, Berlin, Germany
| | - Mit B Bhavsar
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Dmytro Puchkov
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Hannes Maier
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany; Cluster of Excellence "Hearing4all", Hannover, Germany
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Lutze RD, Ingersoll MA, Kelmann RG, Teitz T. Trametinib, a MEK1/2 Inhibitor, Protects Mice from Cisplatin- and Noise-Induced Hearing Loss. Pharmaceuticals (Basel) 2024; 17:735. [PMID: 38931403 PMCID: PMC11206450 DOI: 10.3390/ph17060735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Hearing loss is one of the most common types of disability; however, there is only one FDA-approved drug to prevent any type of hearing loss. Treatment with the highly effective chemotherapy agent, cisplatin, and exposure to high-decibel noises are two of the most common causes of hearing loss. The mitogen-activated protein kinase (MAPK) pathway, a phosphorylation cascade consisting of RAF, MEK1/2, and ERK1/2, has been implicated in both types of hearing loss. Pharmacologically inhibiting BRAF or ERK1/2 is protective against noise- and cisplatin-induced hearing loss in multiple mouse models. Trametinib, a MEK1/2 inhibitor, protects from cisplatin-induced outer hair cell death in mouse cochlear explants; however, to the best of our knowledge, inhibiting MEK1/2 has not yet been shown to be protective against hearing loss in vivo. In this study, we demonstrate that trametinib protects against cisplatin-induced hearing loss in a translationally relevant mouse model and does not interfere with cisplatin's tumor-killing efficacy in cancer cell lines. Higher doses of trametinib were toxic to mice when combined with cisplatin, but lower doses of the drug were protective against hearing loss without any known toxicity. Trametinib also protected mice from noise-induced hearing loss and synaptic damage. This study shows that MEK1/2 inhibition protects against both insults of hearing loss, as well as that targeting all three kinases in the MAPK pathway protects mice from cisplatin- and noise-induced hearing loss.
Collapse
Affiliation(s)
- Richard D. Lutze
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (R.G.K.)
| | - Matthew A. Ingersoll
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (R.G.K.)
| | - Regina G. Kelmann
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (R.G.K.)
| | - Tal Teitz
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (R.G.K.)
- The Scintillon Research Institute, San Diego, CA 92121, USA
| |
Collapse
|
13
|
Maraslioglu-Sperber A, Blanc F, Heller S. Murine cochlear damage models in the context of hair cell regeneration research. Hear Res 2024; 447:109021. [PMID: 38703432 DOI: 10.1016/j.heares.2024.109021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
Understanding the complex pathologies associated with hearing loss is a significant motivation for conducting inner ear research. Lifelong exposure to loud noise, ototoxic drugs, genetic diversity, sex, and aging collectively contribute to human hearing loss. Replicating this pathology in research animals is challenging because hearing impairment has varied causes and different manifestations. A central aspect, however, is the loss of sensory hair cells and the inability of the mammalian cochlea to replace them. Researching therapeutic strategies to rekindle regenerative cochlear capacity, therefore, requires the generation of animal models in which cochlear hair cells are eliminated. This review discusses different approaches to ablate cochlear hair cells in adult mice. We inventoried the cochlear cyto- and histo-pathology caused by acoustic overstimulation, systemic and locally applied drugs, and various genetic tools. The focus is not to prescribe a perfect damage model but to highlight the limitations and advantages of existing approaches and identify areas for further refinement of damage models for use in regenerative studies.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fabian Blanc
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Otolaryngology - Head & Neck Surgery, University Hospital Gui de Chauliac, University of Montpellier, Montpellier, France
| | - Stefan Heller
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
14
|
Lutze RD, Ingersoll MA, Kelmann RG, Teitz T. FDA-Approved MEK1/2 Inhibitor, Trametinib, Protects Mice from Cisplatin and Noise-Induced Hearing Loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595056. [PMID: 38826449 PMCID: PMC11142120 DOI: 10.1101/2024.05.20.595056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Hearing loss is one of the most common types of disability; however, there is only one FDA-approved drug to prevent any type of hearing loss. Treatment with the highly effective chemotherapy agent, cisplatin, and exposure to high decibel noises are two of the most common causes of hearing loss. The mitogen activated protein kinase (MAPK) pathway, a phosphorylation cascade consisting of RAF, MEK1/2, and ERK1/2, has been implicated in both types of hearing loss. Pharmacologically inhibiting BRAF or ERK1/2 is protective from noise and cisplatin-induced hearing loss in multiple mouse models. Trametinib, a MEK1/2 inhibitor, protects from cisplatin induced outer hair cell death in mouse cochlear explants; however, to the best of our knowledge, inhibiting MEK1/2 has not yet been shown to be protective from hearing loss in vivo. In this study, we demonstrate that trametinib protects from cisplatin-induced hearing loss in a translationally relevant mouse model and does not interfere with cisplatin's tumor killing efficacy in cancer cell lines. Higher doses of trametinib were toxic to mice when combined with cisplatin but lower doses of the drug were protective from hearing loss without any known toxicity. Trametinib also protected mice from noise-induced hearing loss and synaptic damage. This study shows that MEK1/2 inhibition protects from both insults of hearing loss and that targeting all three kinases in the MAPK pathway protect from cisplatin and noise-induced hearing loss in mice.
Collapse
Affiliation(s)
- Richard D. Lutze
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Matthew A. Ingersoll
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Regina G. Kelmann
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Tal Teitz
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
15
|
Fujikawa T, Ito T, Okada R, Sawada M, Mohri K, Tateishi Y, Takahashi R, Asakage T, Tsutsumi T. Combined genetic polymorphisms of the GSTT1 and NRF2 genes increase susceptibility to cisplatin-induced ototoxicity: A preliminary study. Hear Res 2024; 445:108995. [PMID: 38518393 DOI: 10.1016/j.heares.2024.108995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVE The genotype-phenotype relationship in cisplatin-induced ototoxicity remains unclear. By assessing early shifts in distortion product otoacoustic emission (DPOAE) levels after initial cisplatin administration, we aimed to discriminate patients' susceptibility to cisplatin-induced ototoxicity and elucidate their genetic background. STUDY DESIGN A prospective cross-sectional study. SETTING Tertiary referral hospital in Japan. PATIENTS Twenty-six patients with head and neck cancer were undergoing chemoradiotherapy with three cycles of 100 mg/m2 cisplatin. INTERVENTIONS Repetitive pure-tone audiometry and DPOAE measurements, and blood sampling for DNA extraction were performed. Patients were grouped into early ototoxicity presence or absence based on whether DPOAE level shifts exceeded the corresponding reference limits of the 21-day test interval. MAIN OUTCOME MEASURES Hearing thresholds after each cisplatin cycle, severity of other adverse events, and polymorphisms in cisplatin-induced ototoxicity-associated genes were compared. RESULTS Early ototoxicity was present in 14 and absent in 12 patients. Ototoxicity presence on DPOAEs was associated with greater progression of hearing loss in frequencies ≥2 kHz throughout therapy and with higher ototoxicity grades compared with ototoxicity absence. Ototoxicity was further associated with grade ≥2 nausea. Ototoxicity presence was genetically associated with the GSTT1 null genotype and G-allele of NFE2L2 rs6721961, whereas ototoxicity absence was associated with the GSTM1 null genotype. Dose-dependent progression of hearing loss was the greatest in the combined genotype pattern of GSTT1 null and the T/G or G/G variants of rs6721961. CONCLUSION Early DPOAE changes reflected genetic vulnerability to cisplatin-induced ototoxicity. Hereditary insufficiency of the antioxidant defense system causes severe cisplatin-induced hearing loss and nausea.
Collapse
Affiliation(s)
- Taro Fujikawa
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan.
| | - Taku Ito
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Ryuhei Okada
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Mitsutaka Sawada
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Kaori Mohri
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Yumiko Tateishi
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Ryosuke Takahashi
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| | - Takeshi Tsutsumi
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo, Tokyo, 113-8510 Japan
| |
Collapse
|
16
|
Chen X, Xiang W, Li L, Xu K. Copper Chaperone Atox1 Protected the Cochlea From Cisplatin by Regulating the Copper Transport Family and Cell Cycle. Int J Toxicol 2024; 43:134-145. [PMID: 37859596 DOI: 10.1177/10915818231206665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Antioxidant 1 copper chaperone (Atox1) may contribute to preventing DDP cochlear damage by regulating copper transport family and cell cycle proteins. A rat model of cochlear damage was developed by placing gelatin sponges treated with DDP in the cochlea. HEI-OC1 cells were treated with 133 μM DDP as a cell model. DDP-induced ototoxicity in rats was confirmed by immunofluorescence (IF) imaging. The damage of DDP to HEI-OC1 cells was assessed by using CCK-8, TUNEL, and flow cytometry. The relationship between Atox1, a member of the copper transport protein family, and the damage to in vivo/vitro models was explored by qRT-PCR, western blot, CCK-8, TUNEL, and flow cytometry. DDP had toxic and other side effects causing cochlear damage and promoted HEI-OC1 cell apoptosis and cell cycle arrest. The over-expression of Atox1 (oe-Atox1) was accomplished by transfecting lentiviral vectors into in vitro/vivo models. We found that oe-Atox1 increased the levels of Atox1, copper transporter 1 (CTR1), and SOD3 in HEI-OC1 cells and decreased the expression levels of ATPase copper transporting α (ATP7A) and ATPase copper transporting β (ATP7B). In addition, the transfection of oe-Atox1 decreased cell apoptosis rate and the number of G2/M stage cells. Similarly, the expression of myosin VI and phalloidin of cochlea cells in vivo decreased. Atox1 ameliorated DDP-induced damage to HEI-OC1 cells or rats' cochlea by regulating the levels of members of the copper transport family.
Collapse
Affiliation(s)
- Xubo Chen
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiren Xiang
- Department of Otolaryngology, Head and Neck Surgery, Jiu Jiang No.1 People's Hospital, Jiujiang, China
| | - Lihua Li
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Xu
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Tan WJT, Vlajkovic SM. Molecular Characteristics of Cisplatin-Induced Ototoxicity and Therapeutic Interventions. Int J Mol Sci 2023; 24:16545. [PMID: 38003734 PMCID: PMC10671929 DOI: 10.3390/ijms242216545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent with proven efficacy in treating various malignancies, including testicular, ovarian, cervical, breast, bladder, head and neck, and lung cancer. Cisplatin is also used to treat tumors in children, such as neuroblastoma, osteosarcoma, and hepatoblastoma. However, its clinical use is limited by severe side effects, including ototoxicity, nephrotoxicity, neurotoxicity, hepatotoxicity, gastrointestinal toxicity, and retinal toxicity. Cisplatin-induced ototoxicity manifests as irreversible, bilateral, high-frequency sensorineural hearing loss in 40-60% of adults and in up to 60% of children. Hearing loss can lead to social isolation, depression, and cognitive decline in adults, and speech and language developmental delays in children. Cisplatin causes hair cell death by forming DNA adducts, mitochondrial dysfunction, oxidative stress, and inflammation, culminating in programmed cell death by apoptosis, necroptosis, pyroptosis, or ferroptosis. Contemporary medical interventions for cisplatin ototoxicity are limited to prosthetic devices, such as hearing aids, but these have significant limitations because the cochlea remains damaged. Recently, the U.S. Food and Drug Administration (FDA) approved the first therapy, sodium thiosulfate, to prevent cisplatin-induced hearing loss in pediatric patients with localized, non-metastatic solid tumors. Other pharmacological treatments for cisplatin ototoxicity are in various stages of preclinical and clinical development. This narrative review aims to highlight the molecular mechanisms involved in cisplatin-induced ototoxicity, focusing on cochlear inflammation, and shed light on potential antioxidant and anti-inflammatory therapeutic interventions to prevent or mitigate the ototoxic effects of cisplatin. We conducted a comprehensive literature search (Google Scholar, PubMed) focusing on publications in the last five years.
Collapse
Affiliation(s)
- Winston J. T. Tan
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand;
- Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| | - Srdjan M. Vlajkovic
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand;
- Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
18
|
Sung CYW, Hayase N, Yuen PS, Lee J, Fernandez K, Hu X, Cheng H, Star RA, Warchol ME, Cunningham LL. Macrophage Depletion Protects Against Cisplatin-Induced Ototoxicity and Nephrotoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567274. [PMID: 38014097 PMCID: PMC10680818 DOI: 10.1101/2023.11.16.567274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cisplatin is a widely used and highly effective anti-cancer drug with significant side effects including ototoxicity and nephrotoxicity. Macrophages, the major resident immune cells in the cochlea and kidney, are important drivers of both inflammatory and tissue repair responses. To investigate the roles of macrophages in cisplatin-induced ototoxicity and nephrotoxicity, we used PLX3397, an FDA-approved inhibitor of the colony-stimulating factor 1 receptor (CSF1R), to eliminate tissue-resident macrophages during the course of cisplatin administration. Mice treated with cisplatin alone (cisplatin/vehicle) had significant hearing loss (ototoxicity) as well as kidney injury (nephrotoxicity). Macrophage ablation using PLX3397 resulted in significantly reduced hearing loss measured by auditory brainstem responses (ABR) and distortion-product otoacoustic emissions (DPOAE). Sensory hair cells in the cochlea were protected against cisplatin-induced death in mice treated with PLX3397. Macrophage ablation also protected against cisplatin-induced nephrotoxicity, as evidenced by markedly reduced tubular injury and fibrosis as well as reduced plasma blood urea nitrogen (BUN) and neutrophil gelatinase-associated lipocalin (NGAL) levels. Mechanistically, our data suggest that the protective effect of macrophage ablation against cisplatin-induced ototoxicity and nephrotoxicity is mediated by reduced platinum accumulation in both the inner ear and the kidney. Together our data indicate that ablation of tissue-resident macrophages represents a novel strategy for mitigating cisplatin-induced ototoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- Cathy Yea Won Sung
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Naoki Hayase
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Peter S.T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - John Lee
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Katharine Fernandez
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Hui Cheng
- Bioinformatics and Biostatistics Collaboration Core, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Mark E. Warchol
- Washington University, Department of Otolaryngology, School of Medicine, Saint Louis, MO
| | - Lisa L. Cunningham
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Kempfle JS, Jung DH. Experimental drugs for the prevention or treatment of sensorineural hearing loss. Expert Opin Investig Drugs 2023; 32:643-654. [PMID: 37598357 DOI: 10.1080/13543784.2023.2242253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
INTRODUCTION Sensorineural hearing loss results in irreversible loss of inner ear hair cells and spiral ganglion neurons. Reduced sound detection and speech discrimination can span all ages, and sensorineural hearing rehabilitation is limited to amplification with hearing aids or cochlear implants. Recent insights into experimental drug treatments for inner ear regeneration and otoprotection have paved the way for clinical trials in order to restore a more physiological hearing experience. Paired with the development of innovative minimally invasive approaches for drug delivery to the inner ear, new, emerging treatments for hearing protection and restoration are within reach. AREAS COVERED This expert opinion provides an overview of the latest experimental drug therapies to protect from and to restore sensorineural hearing loss. EXPERT OPINION The degree and type of cellular damage to the cochlea, the responsiveness of remaining, endogenous cells to regenerative treatments, and the duration of drug availability within cochlear fluids will determine the success of hearing protection or restoration.
Collapse
Affiliation(s)
- Judith S Kempfle
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, UMass Memorial Medical Center, Worcester, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - David H Jung
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Li Y, Zhang T, Song Q, Gao D, Li Y, Jie H, Huang P, Zheng G, Yang J, He J. Cisplatin ototoxicity mechanism and antagonistic intervention strategy: a scope review. Front Cell Neurosci 2023; 17:1197051. [PMID: 37323582 PMCID: PMC10267334 DOI: 10.3389/fncel.2023.1197051] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Cisplatin is a first-line chemotherapeutic agent in the treatment of malignant tumors with remarkable clinical effects and low cost. However, the ototoxicity and neurotoxicity of cisplatin greatly limit its clinical application. This article reviews the possible pathways and molecular mechanisms of cisplatin trafficking from peripheral blood into the inner ear, the toxic response of cisplatin to inner ear cells, as well as the cascade reactions leading to cell death. Moreover, this article highlights the latest research progress in cisplatin resistance mechanism and cisplatin ototoxicity. Two effective protective mechanisms, anti-apoptosis and mitophagy activation, and their interaction in the inner ear are discussed. Additionally, the current clinical preventive measures and novel therapeutic agents for cisplatin ototoxicity are described. Finally, this article also forecasts the prospect of possible drug targets for mitigating cisplatin-induced ototoxicity. These include the use of antioxidants, inhibitors of transporter proteins, inhibitors of cellular pathways, combination drug delivery methods, and other mechanisms that have shown promise in preclinical studies. Further research is needed to evaluate the efficacy and safety of these approaches.
Collapse
Affiliation(s)
- Yingru Li
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Tianyang Zhang
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Qiang Song
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Dekun Gao
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yue Li
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huiqun Jie
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Ping Huang
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Guiliang Zheng
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jun Yang
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jingchun He
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| |
Collapse
|
21
|
Lee DS, Mahal RS, Tharakan T, Cathryn Collopy, Kallogjeri D, Thorstad WL, Adkins DR, Oppelt P, Ley J, Wick CC, Zevallos J. Hearing Outcomes in a Deintensification Trial of Adjuvant Therapy for HPV-Related Oropharyngeal Squamous Cell Carcinoma. Otolaryngol Head Neck Surg 2023; 168:1089-1096. [PMID: 36939390 DOI: 10.1002/ohn.182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/27/2022] [Accepted: 10/08/2022] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To explore whether deintensification of adjuvant therapy reduces ototoxicity among patients with human papillomavirus (HPV)-related oropharyngeal squamous cell carcinoma (OPSCC). STUDY DESIGN Retrospective cohort study. SETTING Single academic center. METHODS The ototoxicity rate among adult patients with HPV-related OPSCC enrolled in the Minimalist Trial (MINT), a prospective phase 2 trial of surgery followed by risk-adjusted deintensified adjuvant therapy (42 Gy radiation given alone or with a single 100 mg/m2 dose of cisplatin), was compared to that among a historical cohort treated with standard adjuvant therapy (60-66 Gy radiation with up to three 100 mg/m2 doses of cisplatin). Ototoxicity was defined as Common Terminology Criteria for Adverse Events v5.0 ≥ Grade 2. Mixed model analysis was performed to investigate the association between deintensified adjuvant therapy and treatment-related hearing loss. RESULTS A total of 29 patients (58 ears) were analyzed in the MINT cohort, and 27 patients (54 ears) in the historical cohort. The ototoxicity rate was 5% (n = 3/58 ears) in the MINT cohort and 46% (n = 25/54 ears) in the historical cohort (difference, 41%; 95% confidence interval [CI] = 27%-56%). Patients in the MINT cohort demonstrated a 95% decrease in risk of ototoxicity compared to those in the historical cohort (adjusted odds ratio: 0.05, 95% CI = 0.01-0.31). Differences in estimated marginal mean threshold shifts were statistically and clinically significant at frequencies ≥ 3 kHz. CONCLUSION The deintensified adjuvant therapy given in MINT led to less ototoxicity than standard adjuvant therapy among patients with HPV-related OPSCC.
Collapse
Affiliation(s)
- David S Lee
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Rajwant S Mahal
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Theresa Tharakan
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Cathryn Collopy
- Department of Otolaryngology-Head and Neck Surgery, Division of Adult Audiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Dorina Kallogjeri
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Wade L Thorstad
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Douglas R Adkins
- Department of Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Peter Oppelt
- Department of Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jessica Ley
- Department of Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Cameron C Wick
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jose Zevallos
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
22
|
Wang X, Zhou Y, Wang D, Wang Y, Zhou Z, Ma X, Liu X, Dong Y. Cisplatin-induced ototoxicity: From signaling network to therapeutic targets. Biomed Pharmacother 2023; 157:114045. [PMID: 36455457 DOI: 10.1016/j.biopha.2022.114045] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/15/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Administration of cisplatin, a common chemotherapeutic drug, has an inevitable side effect of sensorineural hearing loss. The main etiologies are stria vascularis injury, spiral ganglion degeneration, and hair cell death. Over several decades, the research scope of cisplatin-induced ototoxicity has expanded with the discovery of the molecular mechanism mediating inner ear cell death, highlighting the roles of reactive oxygen species and transport channels for cisplatin uptake into inner ear cells. Upon entering hair cells, cisplatin disrupts organelle metabolism, induces oxidative stress, and targets DNA to cause intracellular damage. Recent studies have also reported the role of inflammation in cisplatin-induced ototoxicity. In this article, we preform a narrative review of the latest reported molecular mechanisms of cisplatin-induced ototoxicity, from extracellular to intracellular. We build up a signaling network starting with cisplatin entering into the inner ear through the blood labyrinth barrier, disrupting cochlear endolymph homeostasis, and activating inflammatory responses of the outer hair cells. After entering the hair cells, cisplatin causes hair cell death via DNA damage, redox system imbalance, and mitochondrial and endoplasmic reticulum dysfunction, culminating in programmed cell death including apoptosis, necroptosis, autophagic death, pyroptosis, and ferroptosis. Based on the mentioned mechanisms, prominent therapeutic targets, such as channel-blocking drugs of cisplatin transporter, construction of cisplatin structural analogues, anti-inflammatory drugs, antioxidants, cell death inhibitors, and others, were collated. Considering the recent research efforts, we have analyzed the feasibility of the aforementioned therapeutic strategies and proposed our otoprotective approaches to overcome cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Xilu Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingying Zhou
- Department of Obstetrics & gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dali Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhaoyu Zhou
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiulan Ma
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaofang Liu
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yaodong Dong
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
23
|
Barrallo-Gimeno A, Llorens J. Hair cell toxicology: With the help of a little fish. Front Cell Dev Biol 2022; 10:1085225. [PMID: 36582469 PMCID: PMC9793777 DOI: 10.3389/fcell.2022.1085225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Hearing or balance loss are disabling conditions that have a serious impact in those suffering them, especially when they appear in children. Their ultimate cause is frequently the loss of function of mechanosensory hair cells in the inner ear. Hair cells can be damaged by environmental insults, like noise or chemical agents, known as ototoxins. Two of the most common ototoxins are life-saving medications: cisplatin against solid tumors, and aminoglycoside antibiotics to treat infections. However, due to their localization inside the temporal bone, hair cells are difficult to study in mammals. As an alternative animal model, zebrafish larvae have hair cells similar to those in mammals, some of which are located in a fish specific organ on the surface of the skin, the lateral line. This makes them easy to observe in vivo and readily accessible for ototoxins or otoprotective substances. These features have made possible advances in the study of the mechanisms mediating ototoxicity or identifying new potential ototoxins. Most importantly, the small size of the zebrafish larvae has allowed screening thousands of molecules searching for otoprotective agents in a scale that would be highly impractical in rodent models. The positive hits found can then start the long road to reach clinical settings to prevent hearing or balance loss.
Collapse
Affiliation(s)
- Alejandro Barrallo-Gimeno
- Department de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Campus de Bellvitge, Universitat de Barcelona, L’Hospitalet de Llobregat, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut D'Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
| | - Jordi Llorens
- Department de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Campus de Bellvitge, Universitat de Barcelona, L’Hospitalet de Llobregat, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut D'Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
| |
Collapse
|
24
|
Geohagen B, Zeldin E, Reidy K, Wang T, Gavathiotis E, Fishman YI, LoPachin R, Loeb DM, Weiser DA. Acetophenone protection against cisplatin-induced end-organ damage. Transl Oncol 2022; 27:101595. [PMID: 36477009 PMCID: PMC9723927 DOI: 10.1016/j.tranon.2022.101595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/04/2022] [Accepted: 11/21/2022] [Indexed: 12/09/2022] Open
Abstract
Cisplatin is a widely used and efficacious chemotherapeutic agent for treating solid tumors, yet it causes systemic end-organ damage that is often irreversible and detrimental to quality of life. This includes severe sensorineural hearing loss, hepatotoxicity, and renal injury. Based on the hard-soft acid-base theory, we recently developed two acetophenone-derived, enol-based compounds that directly interfere with the side effects of cisplatin. We investigated organ-specific and generalized toxicity in order to define dose-dependent responses in rodents injected with cisplatin with or without the protective compounds. All metrics that were used as indicators of toxicity showed retention of baseline or control measurements when animals were pre-treated with acetophenones prior to cisplatin administration, while animals injected with no protective compounds showed expected elevations in toxicity measurements or depressions in measurements of organ function. These data support the further investigation of novel acetophenone compounds for the prevention of cisplatin-induced end-organ toxicity.
Collapse
Affiliation(s)
- Brian Geohagen
- Montefiore-Einstein Center for Cancer Care, USA; Montefiore Medical Center, 111 E 210th Street, The Bronx, NY 10467, USA
| | | | - Kimberly Reidy
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, The Bronx, NY 10461, USA; Division of Pediatric Nephrology, Children's Hospital at Montefiore, 3415 Bainbridge Avenue, The Bronx, NY 10467, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, USA
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Albert Einstein College of Medicine, USA; Montefiore-Einstein Center for Cancer Care, USA
| | - Yonatan I Fishman
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, USA
| | - Richard LoPachin
- Department of Anesthesiology, Albert Einstein College of Medicine, USA
| | - David M Loeb
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, USA; Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, The Bronx, NY 10461, USA; Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, USA; Montefiore-Einstein Center for Cancer Care, USA
| | - Daniel A Weiser
- Department of Genetics, Albert Einstein College of Medicine, USA; Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, The Bronx, NY 10461, USA; Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, USA; Montefiore-Einstein Center for Cancer Care, USA.
| |
Collapse
|
25
|
Lee DS, Schrader A, Warchol M, Sheets L. Cisplatin exposure acutely disrupts mitochondrial bioenergetics in the zebrafish lateral-line organ. Hear Res 2022; 426:108513. [PMID: 35534350 PMCID: PMC9745743 DOI: 10.1016/j.heares.2022.108513] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/18/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022]
Abstract
Cisplatin is a commonly used chemotherapeutic agent that causes debilitating high-frequency hearing loss. No targeted therapies currently exist to treat cisplatin ototoxicity, partly because the underlying mechanisms of cisplatin-induced hair cell damage are not completely defined. Zebrafish may offer key insights to cisplatin ototoxicity because their lateral-line organ contains hair cells that are remarkably similar to those within the cochlea but are optically accessible, permitting observation of cisplatin injury in live intact hair cells. In this study, we used a combination of genetically encoded biosensors in zebrafish larvae and fluorescent indicators to characterize changes in mitochondrial bioenergetics in response to cisplatin. Following exposure to cisplatin, confocal imaging of live intact neuromasts demonstrated increased mitochondrial activity. Staining with fixable fluorescent dyes that accumulate in active mitochondria similarly showed hyperpolarized mitochondrial membrane potential. Zebrafish expressing a calcium indicator within their hair cells revealed elevated levels of mitochondrial calcium immediately following completion of cisplatin treatment. A fluorescent ROS indicator demonstrated that these changes in mitochondrial function were associated with increased oxidative stress. After a period of recovery, cisplatin-exposed zebrafish demonstrated caspase-3-mediated apoptosis. Altogether, these findings suggest that cisplatin acutely disrupts mitochondrial bioenergetics and may play a key role in initiating cisplatin ototoxicity.
Collapse
Affiliation(s)
- David S. Lee
- Department of Otolaryngology – Head and Neck Surgery, Washington University School of Medicine, 660 S. Euclid Ave. Campus Box 8115, St. Louis, MO 63110, USA,Corresponding author at: Department of Otolaryngology – Head and Neck Surgery, Washington University School of Medicine, 660 S. Euclid Ave. Campus Box 8115, St. Louis, MO 63110, USA. (D.S. Lee)
| | - Angela Schrader
- Department of Otolaryngology – Head and Neck Surgery, Washington University School of Medicine, 660 S. Euclid Ave. Campus Box 8115, St. Louis, MO 63110, USA
| | - Mark Warchol
- Department of Otolaryngology – Head and Neck Surgery, Washington University School of Medicine, 660 S. Euclid Ave. Campus Box 8115, St. Louis, MO 63110, USA,Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lavinia Sheets
- Department of Otolaryngology – Head and Neck Surgery, Washington University School of Medicine, 660 S. Euclid Ave. Campus Box 8115, St. Louis, MO 63110, USA,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
26
|
Yu W, Zong S, Zhou P, Wei J, Wang E, Ming R, Xiao H. Cochlear Marginal Cell Pyroptosis Is Induced by Cisplatin via NLRP3 Inflammasome Activation. Front Immunol 2022; 13:823439. [PMID: 35529876 PMCID: PMC9067579 DOI: 10.3389/fimmu.2022.823439] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Better understanding the mechanism of cisplatin-induced ototoxicity is of great significance for clinical prevention and treatment of cisplatin-related hearing loss. However, the mechanism of cisplatin-induced inflammatory response in cochlear stria vascularis and the mechanism of marginal cell (MC) damage have not been fully clarified. In this study, a stable model of cisplatin-induced MC damage was established in vitro, and the results of PCR and Western blotting showed increased expressions of NLRP3, Caspase-1, IL-1β, and GSDMD in MCs. Incomplete cell membranes including many small pores appearing on the membrane were also observed under transmission electron microscopy and scanning electron microscopy. In addition, downregulation of NLRP3 by small interfering RNA can alleviate cisplatin-induced MC pyroptosis, and reducing the expression level of TXNIP possesses the inhibition effect on NLRP3 inflammasome activation and its mediated pyroptosis. Taken together, our results suggest that NLRP3 inflammasome activation may mediate cisplatin-induced MC pyroptosis in cochlear stria vascularis, and TXNIP is a possible upstream regulator, which may be a promising therapeutic target for alleviating cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Wenting Yu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shimin Zong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Wei
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Enhao Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruijie Ming
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjun Xiao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Waissbluth S, Maass JC, Sanchez HA, Martínez AD. Supporting Cells and Their Potential Roles in Cisplatin-Induced Ototoxicity. Front Neurosci 2022; 16:867034. [PMID: 35573297 PMCID: PMC9104564 DOI: 10.3389/fnins.2022.867034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cisplatin is a known ototoxic chemotherapy drug, causing irreversible hearing loss. Evidence has shown that cisplatin causes inner ear damage as a result of adduct formation, a proinflammatory environment and the generation of reactive oxygen species within the inner ear. The main cochlear targets for cisplatin are commonly known to be the outer hair cells, the stria vascularis and the spiral ganglion neurons. Further evidence has shown that certain transporters can mediate cisplatin influx into the inner ear cells including organic cation transporter 2 (OCT2) and the copper transporter Ctr1. However, the expression profiles for these transporters within inner ear cells are not consistent in the literature, and expression of OCT2 and Ctr1 has also been observed in supporting cells. Organ of Corti supporting cells are essential for hair cell activity and survival. Special interest has been devoted to gap junction expression by these cells as certain mutations have been linked to hearing loss. Interestingly, cisplatin appears to affect connexin expression in the inner ear. While investigations regarding cisplatin-induced hearing loss have been focused mainly on the known targets previously mentioned, the role of supporting cells for cisplatin-induced ototoxicity has been overlooked. In this mini review, we discuss the implications of supporting cells expressing OCT2 and Ctr1 as well as the potential role of gap junctions in cisplatin-induced cytotoxicity.
Collapse
Affiliation(s)
- Sofia Waissbluth
- Department of Otolaryngology, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Sofia Waissbluth, ;
| | - Juan Cristóbal Maass
- Department of Otolaryngology, Hospital Clínico de la Universidad de Chile, Santiago, Chile
| | - Helmuth A. Sanchez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
28
|
Li J, Liu C, Kaefer S, Youssef M, Zhao B. The Mechanotransduction Channel and Organic Cation Transporter Are Critical for Cisplatin Ototoxicity in Murine Hair Cells. Front Mol Neurosci 2022; 15:835448. [PMID: 35221917 PMCID: PMC8866953 DOI: 10.3389/fnmol.2022.835448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is one of the most widely used chemotherapeutic drugs across the world. However, the serious ototoxic effects, leading to permanent hair cell death and hearing loss, significantly limit the utility of cisplatin. In zebrafish, the functional mechanotransduction channel is required for cisplatin ototoxicity. However, it is still unclear the extent to which the mechanotransduction channel is involved in cisplatin uptake and ototoxicity in mammalian hair cells. Herein, we show that genetically disrupting mechanotransduction in mouse partially protects hair cells from cisplatin-induced hair cell death. Using a fluorescent-dye conjugated cisplatin, we monitored cisplatin uptake in cochlear explants and found that functional mechanotransduction is required for the uptake of cisplatin in murine hair cells. In addition, cimetidine, an inhibitor of the organic cation transporter, also partially protects hair cells from cisplatin ototoxicity. Notably, the otoprotective effects of cimetidine do not require mechanotransduction. These findings suggest that both the mechanotransduction channel and the organic cation transporter are critical for cisplatin ototoxicity in murine hair cells.
Collapse
|
29
|
Yang S, Wu S, Dai W, Pang L, Xie Y, Ren T, Zhang X, Bi S, Zheng Y, Wang J, Sun Y, Zheng Z, Kong J. Tetramethylpyrazine: A Review of Its Antitumor Potential and Mechanisms. Front Pharmacol 2021; 12:764331. [PMID: 34975475 PMCID: PMC8716857 DOI: 10.3389/fphar.2021.764331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer remains a major public health threat. The mitigation of the associated morbidity and mortality remains a major research focus. From a molecular biological perspective, cancer is defined as uncontrolled cell division and abnormal cell growth caused by various gene mutations. Therefore, there remains an urgent need to develop safe and effective antitumor drugs. The antitumor effect of plant extracts, which are characterized by relatively low toxicity and adverse effect, has attracted significant attention. For example, increasing attention has been paid to the antitumor effects of tetramethylpyrazine (TMP), the active component of the Chinese medicine Chuanqiong, which can affect tumor cell proliferation, apoptosis, invasion, metastasis, and angiogenesis, as well as reverse chemotherapeutic resistance in neoplasms, thereby triggering antitumor effects. Moreover, TMP can be used in combination with chemotherapeutic agents to enhance their effects and reduce the side effect associated with chemotherapy. Herein, we review the antitumor effects of TMP to provide a theoretical basis and foundation for the further exploration of its underlying antitumor mechanisms and promoting its clinical application.
Collapse
Affiliation(s)
- Shaojie Yang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuodong Wu
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanlin Dai
- Innovation Institute of China Medical University, Shenyang, China
| | - Liwei Pang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yaofeng Xie
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tengqi Ren
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaolin Zhang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shiyuan Bi
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuting Zheng
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingnan Wang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Sun
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhuyuan Zheng
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Kong
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Jing Kong,
| |
Collapse
|
30
|
Abstract
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 (eg, lead, cadmium, solvents). This article reviews the fundamental mechanisms underlying ototoxicity by clinically relevant, hospital-prescribed medications (ie, 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.
Collapse
Affiliation(s)
- Peter S Steyger
- Translational Hearing Center, Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA.
| |
Collapse
|