1
|
Diepstraten FA, Bertram OMM, Helleman HW, Boerboom RA, van Grotel M, Zsíros J, Tytgat GAM, van Tinteren H, Stokroos RJ, Janssens GO, Hoetink AE, van den Heuvel‐Eibrink MM, Meijer AJM. A Retrospective Evaluation of Ototoxicity Monitoring in a Cohort of Pediatric Patients With Solid Tumors, Treated in the Dutch National Cancer Center. Cancer Rep (Hoboken) 2024; 7:e70046. [PMID: 39589176 PMCID: PMC11590333 DOI: 10.1002/cnr2.70046] [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: 05/04/2024] [Revised: 09/22/2024] [Accepted: 10/03/2024] [Indexed: 11/27/2024] Open
Abstract
INTRODUCTION Ototoxicity is an adverse effect of childhood cancer treatment with a negative impact on speech-language development and quality of life. This study aimed to retrospectively assess ototoxicity monitoring in a national cohort of pediatric patients with solid tumors, examining the frequency and determinants associated with hearing loss (HL). METHODS This retrospective cohort study included 305 patients treated between 2015 and 2020 at the Princess Máxima Center. Patients receiving platinum agents, head and neck radiotherapy, and/or ear-nose-throat surgery were analyzed. Electronic patient files provided demographic, clinical, and audiological data. HL was defined as Muenster ≥ 2b or SIOP ≥ 2 grade. Associations between clinical characteristics and HL occurrence were analyzed using logistic regression analysis. RESULTS Audiological monitoring was performed at baseline (62.6%), during treatment (79.0%), and at the end of treatment (82.1%). Post treatment, 51.2% and 36.5% experienced Muenster and SIOP-defined HL, respectively. Multivariable analyses revealed that age at diagnosis (OR 0.9, 95% CI 0.9-1.0), total cumulative dose cisplatin per 100 mg/m2 (OR 1.6, 95% CI 1.4-2.0), and vincristine treatment (OR 3.3, 95% CI 1.4-7.8) remained significantly associated with Muenster grade ≥ 2b HL. Age at diagnosis in years (OR 0.9, 95% CI 0.8-1.0), total cumulative dose cisplatin per 100 mg/m2 (OR 1.5, 95% CI 1.2-1.8), and male sex (OR 2.7, 95% CI 1.4-5.3) were associated with SIOP ≥ 2 HL. CONCLUSION This study shows that more than half of the children treated with ototoxic cancer therapies develop HL by the end of treatment. Therefore, audiological monitoring during and after treatment is essential. Improved insight into clinical determinants aids in identifying patients at high risk for HL, who may benefit from prevention strategies that are currently being implemented.
Collapse
Affiliation(s)
| | | | - Hiske W. Helleman
- Department of AudiologyUniversity Medical Center UtrechtUtrechtNetherlands
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity Medical Center UtrechtUtrechtNetherlands
| | - Ralf A. Boerboom
- Department of AudiologyUniversity Medical Center UtrechtUtrechtNetherlands
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity Medical Center UtrechtUtrechtNetherlands
| | | | - József Zsíros
- Princess Máxima Center for Pediatric OncologyUtrechtNetherlands
| | | | | | - Robert J. Stokroos
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity Medical Center UtrechtUtrechtNetherlands
- UMC Brain CenterUniversity Medical Center UtrechtUtrechtNetherlands
| | - Geert O. Janssens
- Princess Máxima Center for Pediatric OncologyUtrechtNetherlands
- Department of Radiation OncologyUniversity Medical Center UtrechtUtrechtNetherlands
| | - Alex E. Hoetink
- Department of AudiologyUniversity Medical Center UtrechtUtrechtNetherlands
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity Medical Center UtrechtUtrechtNetherlands
| | - Marry M. van den Heuvel‐Eibrink
- Princess Máxima Center for Pediatric OncologyUtrechtNetherlands
- University Medical Center Utrecht‐Wilhelmina Children's HospitalUtrechtNetherlands
| | | |
Collapse
|
2
|
Miao DNR, Wilke MAP, Pham J, Ladha F, Singh M, Arsenio J, Luca E, Dabdoub A, Yang W, Yang JJ, Drögemöller BI. Leveraging large-scale datasets and single cell omics data to develop a polygenic score for cisplatin-induced ototoxicity. Hum Genomics 2024; 18:112. [PMID: 39380081 PMCID: PMC11463131 DOI: 10.1186/s40246-024-00679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Cisplatin-induced ototoxicity (CIO), characterized by irreversible and progressive bilateral hearing loss, is a prevalent adverse effect of cisplatin chemotherapy. Alongside clinical risk factors, genetic variants contribute to CIO and genome-wide association studies (GWAS) have highlighted the polygenicity of this adverse drug reaction. Polygenic scores (PGS), which integrate information from multiple genetic variants across the genome, offer a promising tool for the identification of individuals who are at higher risk for CIO. Integrating large-scale hearing loss GWAS data with single cell omics data holds potential to overcome limitations related to small sample sizes associated with CIO studies, enabling the creation of PGSs to predict CIO risk. RESULTS We utilized a large-scale hearing loss GWAS and murine inner ear single nuclei RNA-sequencing (snRNA-seq) data to develop two polygenic scores: a hearing loss PGS (PGSHL) and a biologically informed PGS for CIO (PGSCIO). The PGSCIO included only variants which mapped to genes that were differentially expressed within cochlear cells that showed differential abundance in the murine snRNA-seq data post-cisplatin treatment. Evaluation of the association of these PGSs with CIO in our target CIO cohort revealed that PGSCIO demonstrated superior performance (P = 5.54 × 10- 5) relative to PGSHL (P = 2.93 × 10- 3). PGSCIO was also associated with CIO in our test cohort (P = 0.04), while the PGSHL did not show a significant association with CIO (P = 0.52). CONCLUSION This study developed the first PGS for CIO using a large-scale hearing loss dataset and a biologically informed filter generated from cisplatin-treated murine inner ear snRNA-seq data. This innovative approach offers new avenues for developing PGSs for pharmacogenomic traits, which could contribute to the implementation of tailored therapeutic interventions. Further, our approach facilitated the identification of specific cochlear cells that may play critical roles in CIO. These novel insights will guide future research aimed at developing targeted therapeutic strategies to prevent CIO.
Collapse
Affiliation(s)
- Deanne Nixie R Miao
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - MacKenzie A P Wilke
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - John Pham
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Feryal Ladha
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mansumeet Singh
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Janilyn Arsenio
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Emilia Luca
- Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Wejian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- CancerCare Manitoba Research Institute, Winnipeg, MB, Canada.
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Centre of Aging, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
3
|
de Bont JM, Schouten-van Meeteren AYN. Long-term quality of survival after pediatric low-grade glioma. Childs Nerv Syst 2024; 40:3341-3355. [PMID: 39400717 DOI: 10.1007/s00381-024-06631-1] [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: 04/23/2024] [Accepted: 09/21/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Low-grade glioma is the most common brain tumor in children with different modes of treatment and a high overall survival. Low-grade glioma is considered a chronic disease, since residual tumor is present in many children. The tumor and its treatment lead to acquired brain injury with diverse consequences for later life based on factors like the diverse tumor locations, treatment(s) applied, neurofibromatosis type 1, and age at diagnosis. METHODS An overview of affected domains is provided based upon cohort studies from literature and partially based on clinical experience with a practical approach regarding each domain of functioning in order to provide insight in the requirements for long-term care assistance after childhood low-grade glioma. RESULTS The diverse domains that can potentially be affected are described as follows: motor function, speech, eating and swallowing, sensory functions, seizures, neuropathy, organ function after systemic treatment, late effects due to cranial radiation (vascular changes and secondary tumors, endocrine and hypothalamic function, sleep and energy, neuro-cognition and education, psychosocial effects, and quality of life. CONCLUSION Insight in affected domains guides advices for medical follow-up, diagnostics, supportive instructions, and assistive measures per domain of functioning and provide insight in the requirements for long-term care assistance after childhood low-grade glioma.
Collapse
Affiliation(s)
- Judith M de Bont
- Department Late Effects Clinic, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, Netherlands
| | | |
Collapse
|
4
|
Li J, Rouse SL, Matthews IR, Park Y, Eltawil Y, Sherr EH, Chan DK. Modulating the unfolded protein response with ISRIB mitigates cisplatin ototoxicity. Sci Rep 2024; 14:22382. [PMID: 39333235 PMCID: PMC11437005 DOI: 10.1038/s41598-024-70561-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/19/2024] [Indexed: 09/29/2024] Open
Abstract
Cisplatin is a commonly used chemotherapy agent with a nearly universal side effect of sensorineural hearing loss. The cellular mechanisms underlying cisplatin ototoxicity are poorly understood. Efforts in drug development to prevent or reverse cisplatin ototoxicity have largely focused on pathways of oxidative stress and apoptosis. An effective treatment for cisplatin ototoxicity, sodium thiosulfate (STS), while beneficial when used in standard risk hepatoblastoma, is associated with reduced survival in disseminated pediatric malignancy, highlighting the need for more specific drugs without potential tumor protective effects. The unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways have been shown to be involved in the pathogenesis of noise-induced hearing loss and cochlear synaptopathy in vivo, and these pathways have been implicated broadly in cisplatin cytotoxicity. This study sought to determine whether the UPR can be targeted to prevent cisplatin ototoxicity. Neonatal cochlear cultures and HEK cells were exposed to cisplatin, and UPR marker gene expression and cell death measured. Treatment with ISRIB (Integrated Stress Response InhIBitor), a drug that activates eif2B and downregulates the pro-apoptotic PERK/CHOP pathway of the UPR, was tested for its ability to reduce apoptosis in HEK cells, hair-cell death in cochlear cultures, and hearing loss using an in vivo mouse model of cisplatin ototoxicity. Finally, to evaluate whether ISRIB might interfere with cisplatin chemoeffectiveness, we tested it in head and neck squamous cell carcinoma (HNSCC) cell-based assays of cisplatin cytotoxicity. Cisplatin exhibited a biphasic, non-linear dose-response of cell death and apoptosis that correlated with different patterns of UPR marker gene expression in HEK cells and cochlear cultures. ISRIB treatment protected against cisplatin-induced hearing loss and hair-cell death, but did not impact cisplatin's cytotoxic effects on HNSCC cell viability, unlike STS. These findings demonstrate that targeting the pro-apoptotic PERK/CHOP pathway with ISRIB can mitigate cisplatin ototoxicity without reducing anti-cancer cell effects, suggesting that this may be a viable strategy for drug development.
Collapse
Affiliation(s)
- Jiang Li
- Department of Neurology, UCSF, San Francisco, USA
| | - Stephanie L Rouse
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco (UCSF), 513 Parnassus Ave, Rm 719, San Francisco, CA, 94143, USA
- Department of Neurobiology, Harvard Medical School, Boston, USA
| | - Ian R Matthews
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco (UCSF), 513 Parnassus Ave, Rm 719, San Francisco, CA, 94143, USA
| | - Yesai Park
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco (UCSF), 513 Parnassus Ave, Rm 719, San Francisco, CA, 94143, USA
| | - Yasmin Eltawil
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco (UCSF), 513 Parnassus Ave, Rm 719, San Francisco, CA, 94143, USA
| | - Elliott H Sherr
- Department of Neurology, UCSF, San Francisco, USA
- Department of Pediatrics, Institute of Human Genetics, Weill Institute for Neurosciences, UCSF, San Francisco, USA
| | - Dylan K Chan
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco (UCSF), 513 Parnassus Ave, Rm 719, San Francisco, CA, 94143, USA.
| |
Collapse
|
5
|
Davis ME, Guarini E, O'Connor K, Francis JH, Abramson DH. Hearing Evaluations in Children With Retinoblastoma Treated With Intra-arterial Carboplatin Chemotherapy: A Single Institution Review. J Pediatr Ophthalmol Strabismus 2024:1-6. [PMID: 39254185 DOI: 10.3928/01913913-20240807-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE To determine whether the administration of intra-arterial carboplatin affected the hearing of children with retinoblastoma. METHODS Children with retinoblastoma who were treated with intra-arterial carboplatin chemotherapy were included. Hearing tests before chemotherapy including tympanometry, distortion product otoacoustic emissions, and audiogram (if achievable) were performed and repeated 3 to 9 months after concluding intra-arterial therapy. The study was approved by the institutional review board. Patients were identified from the retinoblastoma clinic when the treatment plan included intra-arterial carboplatin chemotherapy. Children were excluded if they had previous intra-arterial carboplatin or preexisting hearing loss but were included if they had systemic carboplatin and dosing was available. Tympanometry was performed to rule out inner ear fluid. All examinations were performed by a certified audiologist with the same equipment, calibrated regularly by a certified technician. RESULTS Twenty-two children (32 eyes) were evaluable. Because most children are diagnosed at a young age and are unable to participate in an audiogram, distortion product otoacoustic emission measurement was the primary measurement. No child displayed hearing loss. CONCLUSIONS Intra-arterial chemotherapy with carboplatin did not cause ototoxicity in any child by distortion product otoacoustic emission measurement in contrast to systemic chemotherapy where ototoxicity is common. Distortion product otoacoustic emission levels were essentially unchanged from before to after intra-arterial chemotherapy in children with retinoblastoma. These findings suggest that intra-arterial carboplatin does not affect outer hair cell function, and distortion product otoacoustic emission tests can provide useful information when monitoring children at risk of developing carboplatin ototoxicity. [J Pediatr Ophthalmol Strabismus. 20XX;X(X):XXX-XXX.].
Collapse
|
6
|
Li J, Rouse SL, Matthews IR, Sherr EH, Chan DK. Modulating the Unfolded Protein Response with ISRIB Mitigates Cisplatin Ototoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.17.562797. [PMID: 37905009 PMCID: PMC10614842 DOI: 10.1101/2023.10.17.562797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cisplatin is a commonly used chemotherapy agent with a nearly universal side effect of sensorineural hearing loss. The cellular mechanisms underlying cisplatin ototoxicity are poorly understood. Efforts in drug development to prevent or reverse cisplatin ototoxicity have largely focused on pathways of oxidative stress and apoptosis. An effective treatment for cisplatin ototoxicity, sodium thiosulfate (STS), while beneficial when used in standard risk hepatoblastoma, is associated with reduced survival in disseminated pediatric malignancies, highlighting the need for more specific drugs without potential tumor protective effects. The unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways have been shown to be involved in the pathogenesis of noise-induced hearing loss and cochlear synaptopathy in vivo, and these pathways have been implicated broadly in cisplatin cytotoxicity. This study sought to determine whether the UPR can be targeted to prevent cisplatin ototoxicity. Neonatal cochlear cultures and HEK cells were exposed to cisplatin and UPR-modulating drugs, and UPR marker gene expression and cell death measured. Treatment with ISRIB, a drug that activates eif2B and downregulates the pro-apoptotic PERK/CHOP pathway of the UPR, was tested in an in vivo mouse model of cisplatin ototoxicity and well as a head and neck squamous cell carcinoma (HNSCC) cell-based assay of cisplatin cytotoxicity. Cisplatin exhibited a biphasic, non-linear dose-response of cell death and apoptosis that correlated with different patterns of UPR marker gene expression in HEK cells and cochlear cultures. ISRIB treatment protected against cisplatin-induced hearing loss and hair-cell death, but did not impact the cytotoxic effects of cisplatin on HNSCC cell viability, unlike STS. These findings demonstrate that targeting the pro-apoptotic PERK/CHOP pathway with ISRIB can mitigate cisplatin ototoxicity without reducing anti-cancer cell effects, suggesting that this may be a viable strategy for drug development.
Collapse
|
7
|
Funt SA, Knezevic A, Wilson K, Bromberg M, Budnick A, O’Connor KL, McHugh DJ, Larsen E, Bajorin DF, Motzer RJ, Tonorezos ES, Patil S, Feldman DR. Ototoxicity associated with high-dose carboplatin for patients with previously treated germ cell tumors. Cancer 2023; 129:3952-3961. [PMID: 37715631 PMCID: PMC11305123 DOI: 10.1002/cncr.34991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/26/2023] [Accepted: 07/18/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND High-dose carboplatin is an essential part of curative high-dose chemotherapy (HDCT) for patients with previously treated germ cell tumors (GCTs). Although hearing loss (HL) is a known side effect of HDCT, data on its severity and characteristics are limited. METHODS Eligible patients received HDCT for GCTs from 1993 to 2017 and had audiograms before and after HDCT. HL severity was classified by American Speech-Language-Hearing Association criteria, and mean change in hearing threshold at each frequency (0.25-8 kHz) was estimated from pre- to post-HDCT and between HDCT cycles. RESULTS Of 115 patients (median age, 32 years), 102 (89%) received three cycles of HDCT. Of 106 patients with normal hearing to mild HL in the speech frequencies (0.5-4 kHz) before HDCT, 70 (66%) developed moderate to profound HL in the speech frequencies after HDCT. Twenty-five patients (22%) were recommended for hearing aids after HDCT. Patients with moderate to profound HL isolated to the higher frequencies (6-8 kHz) before HDCT were more likely to develop moderate to profound HL in the speech frequencies after HDCT (94% vs. 61%; p = .01) and to be recommended for hearing aids (39% vs. 18%; p = .05). CONCLUSIONS HL was frequent after HDCT for GCTs, with most patients developing at least moderate HL in the speech frequencies and approximately one in five recommended for hearing aids. Moderate to profound HL isolated to high frequencies at baseline was predictive of more clinically significant hearing impairment after HDCT. PLAIN LANGUAGE SUMMARY Some patients with germ cell tumors, the most common malignancy in adolescent and young adult men, are not cured with standard-dose chemotherapy and require high-dose chemotherapy (HDCT). Using detailed hearing assessments of patients receiving HDCT, we found that most patients developed significant hearing loss and that one in five needed hearing aids. Thus, strategies to reduce this side effect are urgently needed, and all patients receiving HDCT should have a hearing test after therapy.
Collapse
Affiliation(s)
- Samuel A. Funt
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Kaamilah Wilson
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Maria Bromberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Amy Budnick
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Kerri L. O’Connor
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Deaglan J. McHugh
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Erik Larsen
- Formerly of Decibel Therapeutics, Boston, MA, USA
| | - Dean F. Bajorin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Robert J. Motzer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | - Sujata Patil
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Biostatistics, Cleveland Clinic, Cleveland, Ohio, USA
| | - Darren R. Feldman
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
8
|
Scott EN, Joseph AA, Dhanda A, Tanoshima R, Brooks B, Rassekh SR, Ross CJD, Carleton BC, Loucks CM. Systematic Critical Review of Genetic Factors Associated with Cisplatin-induced Ototoxicity: Canadian Pharmacogenomics Network for Drug Safety 2022 Update. Ther Drug Monit 2023; 45:714-730. [PMID: 37726872 DOI: 10.1097/ftd.0000000000001113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/01/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Cisplatin is commonly used to treat solid tumors; however, its use can be complicated by drug-induced hearing loss (ie, ototoxicity). The presence of certain genetic variants has been associated with the development/occurrence of cisplatin-induced ototoxicity, suggesting that genetic factors may be able to predict patients who are more likely to develop ototoxicity. The authors aimed to review genetic associations with cisplatin-induced ototoxicity and discuss their clinical relevance. METHODS An updated systematic review was conducted on behalf of the Canadian Pharmacogenomics Network for Drug Safety, based on the Preferred Reporting Items for Systematic reviews and Meta-Analyses 2020 statement. Pharmacogenomic studies that reported associations between genetic variation and cisplatin-induced ototoxicity were included. The evidence on genetic associations was summarized and evaluated, and knowledge gaps that can be used to inform future pharmacogenomic studies identified. RESULTS Overall, 40 evaluated reports, considering 47 independent patient populations, captured associations involving 24 genes. Considering GRADE criteria, genetic variants in 2 genes were strongly (ie, odds ratios ≥3) and consistently (ie, replication in ≥3 independent populations) predictive of cisplatin-induced ototoxicity. Specifically, an ACYP2 variant has been associated with ototoxicity in both children and adults, whereas TPMT variants are relevant in children. Encouraging evidence for associations involving several other genes also exists; however, further research is necessary to determine potential clinical relevance. CONCLUSIONS Genetic variation in ACYP2 and TPMT may be helpful in predicting patients at the highest risk of developing cisplatin-induced ototoxicity. Further research (including replication studies considering diverse pediatric and adult patient populations) is required to determine whether genetic variation in additional genes may help further identify patients most at risk.
Collapse
Affiliation(s)
- Erika N Scott
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Akshaya A Joseph
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
| | - Angie Dhanda
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
| | - Reo Tanoshima
- Department of Pediatrics, Yokohama City University Hospital, Yokohama, Japan
- YCU Center for Novel and Exploratory Clinical Trials, Yokohama City University Hospital, Yokohama, Japan
| | - Beth Brooks
- Audiology and Speech Pathology Department, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
- School of Audiology and Speech Science, UBC, Vancouver, British Columbia, Canada
| | - S Rod Rassekh
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Oncology, Hematology and Bone Marrow Transplant, British Columbia Children's Hospital and UBC, Vancouver, British Columbia, Canada
| | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, UBC, Vancouver, British Columbia, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
- Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - Catrina M Loucks
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
| |
Collapse
|
9
|
Hu S, Sun Q, Xu F, Jiang N, Gao J. Age-related hearing loss and its potential drug candidates: a systematic review. Chin Med 2023; 18:121. [PMID: 37730634 PMCID: PMC10512576 DOI: 10.1186/s13020-023-00825-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Age-related hearing loss (ARHL) is one of the main illnesses afflicting the aged population and has a significant negative impact on society, economy, and health. However, there is presently no appropriate therapeutic treatment of ARHL due to the absence of comprehensive trials. OBJECTIVES The goal of this review is to systematically evaluate and analyze recent statistics on the pathologic classifications, risk factors, treatment strategies, and drug candidates of ARHL, including that from traditional Chinese medicine (TCM), to provide potential new approaches for preventing and treating ARHL. METHODS Literature related to ARHL was conducted in databases such as PubMed, WOS, China National Knowledge Infrastructure (CNKI), and Wanfang from the establishment of the database to Jan, 2023. The pathology, causal factor, pathophysiological mechanism, treatment strategy, and the drug candidate of ARHL were extracted and pooled for synthesis. RESULTS Many hypotheses about the etiology of ARHL are based on genetic and environmental elements. Most of the current research on the pathology of ARHL focuses on oxidative damage, mitochondrial dysfunction, inflammation, cochlear blood flow, ion homeostasis, etc. In TCM, herbs belonging to the kidney, lung, and liver meridians exhibit good hearing protection. Seven herbs belonging to the kidney meridian, 9 belonging to the lung meridian, and 4 belonging to the liver meridian were ultimately retrieved in this review, such as Polygonum multiflorum Thunb., Panax ginseng C.A. Mey, and Pueraria lobata (Willd.) Ohwi. Their active compounds, 2,3,4',5-Tetrahydroxystilbene-2-O-D-glucoside, ginsenoside Rb1, and puerarin, may act as the molecular substance for their anti-ARHL efficacy, and show anti-oxidative, neuroprotective, anti-inflammatory, anti-apoptotic, or mitochondrial protective effects. CONCLUSION Anti-oxidants, modulators of mitochondrial function, anti-inflammation agents, vasodilators, K+ channel openers, Ca2+ channel blockers, JNK inhibitors, and nerve growth factors/neurotrophic factors all contribute to hearing protection, and herbs are an important source of potential anti-ARHL drugs.
Collapse
Affiliation(s)
- Shiyu Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Qingru Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Fei Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Ninghua Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| |
Collapse
|
10
|
Du K, Liao P, Yang S, von Trentini D, Sharma K, Shi X, Murray CB, Li D, Dmochowski IJ. Chelate-functionalized magnetic micelles for sequestration of cisplatin. NANOSCALE ADVANCES 2023; 5:3955-3963. [PMID: 37496616 PMCID: PMC10367963 DOI: 10.1039/d3na00290j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/02/2023] [Indexed: 07/28/2023]
Abstract
Many cancer patients suffer permanent hearing loss due to accumulation of ototoxic cisplatin in the inner ear. In this study, two types of 100 nm magnetic micelles were developed to sequester cisplatin from aqueous solutions, with the goal of eliminating cochlear ototoxins via magnetic microsurgery. The micellar surface was quantitatively functionalized with anionic S-rich ligands and the micelle core encapsulated superparamagnetic iron oxide nanoparticles. Exceptionally effective sequestration is demonstrated, with removal of greater than 95 and 50% of solution Pt, by means of centrifugal filtration and magnetic extraction. Attraction between negatively charged micellar surfaces and cationic Pt-species played a critical role and was only partially screened by physiologic salt solution. Importantly, magnetic micelles introduce negligible impact on the integrity of inner ear hair cells, demonstrating excellent biocompatibility. This study showcases successful magnetic sequestration of Pt-based ototoxins using highly applicable nano-micellar materials. More generally, these examples highlight features of the micelle-water interfacial environment that are important in developing nanomaterials for metallo-medicinal applications.
Collapse
Affiliation(s)
- Kang Du
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| | - Pan Liao
- Department of Otorhinolaryngology, University of Pennsylvania Philadelphia PA 19104 USA
| | - Shengsong Yang
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| | - Dora von Trentini
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| | - Kushal Sharma
- Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University Portland OR 97239 USA
| | - Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University Portland OR 97239 USA
| | - Christopher B Murray
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
- Department of Materials Science and Engineering, University of Pennsylvania Philadelphia PA 19104 USA
| | - Daqing Li
- Department of Otorhinolaryngology, University of Pennsylvania Philadelphia PA 19104 USA
| | - Ivan J Dmochowski
- Department of Chemistry, University of Pennsylvania Philadelphia PA 19104 USA
| |
Collapse
|
11
|
Omar NE, Elewa H. Cisplatin-induced ototoxicity: a novel approach to an ancient problem. Pharmacogenet Genomics 2023; 33:111-115. [PMID: 37068004 DOI: 10.1097/fpc.0000000000000497] [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: 04/18/2023]
Abstract
With the scarcity of pharmacological otoprotective agents against cisplatin-induced ototoxicity (CIO), researchers find themselves compelled to look at and navigate all possible strategies to identify ways to prevent CIO. One of these promising strategies is pharmacogenomic implementation. This strategy aims for identifying and detecting high-risk genetic variants to tailor cisplatin therapy to reach the best survival outcomes with the least risk of ototoxicity.
Collapse
Affiliation(s)
- Nabil E Omar
- Pharmacy Department, National Center for Cancer Care and Research, Hamad Medical Corporation
- Clinical and Population Health Research, College of Pharmacy, Qatar University, Doha, Qatar
| | - Hazem Elewa
- Clinical and Population Health Research, College of Pharmacy, Qatar University, Doha, Qatar
| |
Collapse
|
12
|
Brock P, Meijer A, Kogner P, Ansari M, Capra M, Geller J, Heuvel-Eibrink MVD, Knight K, Kruger M, Lindemulder S, Maibach R, O'Neill A, Papadakis V, Rajput K, Bleyer A, Bouffet E, Sullivan M. Sodium thiosulfate as cisplatin otoprotectant in children: The challenge of when to use it. Pediatr Blood Cancer 2023; 70:e30248. [PMID: 36772889 DOI: 10.1002/pbc.30248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 02/12/2023]
Affiliation(s)
- Penelope Brock
- Department of Paediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Annelot Meijer
- Department of Pediatric Oncology, Prinses Maxima Centrum, Utrecht, The Netherlands
| | - Per Kogner
- Department of Pediatric Oncology and Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, Sweden
| | - Marc Ansari
- Pediatric Hematology and Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Michael Capra
- Paediatric Oncology, Our Lady's Children's Hospital, Dublin, Ireland
| | - James Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Kristin Knight
- Department of Pediatric Audiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Mariana Kruger
- Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, South Africa
| | - Susan Lindemulder
- Department of Pediatric Oncology, Oregon Health and Science University, Portland, Oregon, USA
| | - Rudolf Maibach
- Department of Statistics, International Breast Cancer Study Group, Bern, Switzerland
| | - Allison O'Neill
- Children's Cancer and Blood Disorders Center, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Vassilios Papadakis
- Department of Pediatric Oncology, Agia Sofia Children's Hospital, Athens, Greece
| | - Kaukab Rajput
- Department of Paediatric Audiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Archie Bleyer
- Radiation Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Eric Bouffet
- Division of Pediatric Neuro-Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Sullivan
- Children's Cancer Centre and Department of Paediatric Oncology, Royal Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Du P, Liu T, Luo P, Li H, Tang W, Zong S, Xiao H. SIRT3/GLUT4 signaling activation by metformin protect against cisplatin-induced ototoxicity in vitro. Arch Toxicol 2023; 97:1147-1162. [PMID: 36800006 DOI: 10.1007/s00204-023-03457-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023]
Abstract
Cisplatin is highly effective for killing tumor cells. However, as one of its side effects, ototoxicity limits the clinical application of cisplatin. The mechanisms of cisplatin-induced ototoxicity have not been fully clarified yet. SIRT3 is a deacetylated protein mainly located in mitochondria, which regulates a variety of physiological processes in cells. The role of SIRT3 in cisplatin-induced hair cell injury has not been founded. In this study, primary cultured cochlear explants exposed to 5 μM cisplatin, as well as OC-1 cells exposed to 10 μM cisplatin, were used to establish models of cisplatin-induced ototoxicity in vitro. We found that when combined with cisplatin, metformin (75 μM) significantly up-regulated the expression of SIRT3 and alleviated cisplatin-induced apoptosis of hair cells. We regulated the expression of SIRT3 to explore the role of SIRT3 in cisplatin-induced auditory hair cell injury. Overexpression of SIRT3 promoted the survival of auditory hair cells and alleviated the apoptosis of auditory hair cells. In contrast, knockdown of SIRT3 impaired the protective effect of metformin and exacerbated cisplatin injury. In addition, we found that the protective effect of SIRT3 may be achieved by regulating GLUT4 translocation and rescuing impaired glucose uptake caused by cisplatin. Our study confirmed that upregulation of SIRT3 may antagonize cisplatin-induced ototoxicity, and provided a new perspective for the study of cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Peiyu Du
- Department of Otolaryngology-Head and Neck Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tianyi Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pan Luo
- Department of Otolaryngology-Head and Neck Surgery, Wuhan Central Hospital, Wuhan, China
| | - Hejie Li
- Department of Otolaryngology-Head and Neck Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Tang
- Department of Otolaryngology-Head and Neck Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shimin Zong
- Department of Otolaryngology-Head and Neck Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Hongjun Xiao
- Department of Otolaryngology-Head and Neck Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Role of Cisplatin Dose Intensity and TPMT Variation in the Development of Hearing Loss in Children. Ther Drug Monit 2023; 45:345-353. [PMID: 36917731 DOI: 10.1097/ftd.0000000000001085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/30/2022] [Indexed: 03/15/2023]
Abstract
BACKGROUND Cisplatin, widely used in the treatment of solid tumors, causes permanent hearing loss in more than 60% of treated children. Previous studies have implicated several clinical factors in the development of ototoxicity, including cumulative cisplatin dose. However, the role of cisplatin dose intensity in the development of hearing loss in children remains unclear. Pharmacogenetic studies have also identified genetic variants in TPMT that increase the risk of cisplatin-induced hearing loss. This study aims to determine whether cisplatin dose intensity contributes to the risk of hearing loss in children and whether genetic variations in TPMT further modifies the risk of cisplatin-induced hearing loss. METHODS The authors genotyped 371 cisplatin-treated children for the presence of any 3 TPMT-risk variants. Patients were categorized into high-, moderate-, and low-intensity cisplatin dosing groups according to the cisplatin dose administered per unit time. Kaplan-Meier curves were plotted to compare the cumulative incidence of hearing loss between the genotype and dose intensity groups. RESULTS Patients receiving cisplatin at high dose intensity experienced significantly higher incidences of ototoxicity than those receiving cisplatin at low dose intensity (P = 9 × 10-7). Further stratification by TPMT genotype revealed that carriers of ≥1 TPMT variants receiving high-intensity cisplatin developed ototoxicity sooner and more often than their wild-type counterparts (93.8% vs. 56.6% at 12 months; P = 5 × 10-5) and noncarriers receiving low-intensity cisplatin (21.2% at 12 months). CONCLUSIONS Cisplatin dose intensity is strongly associated with ototoxicity development in children, and this risk is further increased by the presence of TPMT-risk alleles.
Collapse
|
15
|
Hong DZ, Ong TCC, Timbadia DP, Tan HTA, Kwa ED, Chong WQ, Goh BC, Loh WS, Loh KS, Tan EC, Tay JK. Systematic Review and Meta-Analysis of the Influence of Genetic Variation on Ototoxicity in Platinum-Based Chemotherapy. Otolaryngol Head Neck Surg 2023; 168:1324-1337. [PMID: 36802061 DOI: 10.1002/ohn.222] [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: 05/03/2022] [Revised: 11/04/2022] [Accepted: 11/19/2022] [Indexed: 02/19/2023]
Abstract
OBJECTIVE The objective of this meta-analysis is to evaluate the impact of genetic polymorphisms on platinum-based chemotherapy (PBC)-induced ototoxicity. DATA SOURCES Systematic searches of PubMed, Embase, Cochrane, and Web of Science were conducted from the inception of the databases to May 31, 2022. Abstracts and presentations from conferences were also reviewed. REVIEW METHODS Four investigators independently extracted data in adherence to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Differences in the prevalence of PBC-induced ototoxicity between reference and variant (i) genotypes and (ii) alleles were analyzed. The overall effect size was presented using the random-effects model as an odds ratio (OR) with a 95% confidence interval (CI). RESULTS From 32 included articles, 59 single nucleotide polymorphisms on 28 genes were identified, with 4406 total unique participants. For allele frequency analysis, the A allele in ACYP2 rs1872328 was positively associated with ototoxicity (OR: 2.61; 95% CI: 1.06-6.43; n = 2518). Upon limiting to cisplatin use only, the T allele of COMT rs4646316 and COMT rs9332377 revealed significant results. For genotype frequency analysis, the CT/TT genotype in ERCC2 rs1799793 demonstrated an otoprotective effect (OR: 0.50; 95% CI: 0.27-0.94; n = 176). Excluding studies using carboplatin or concomitant radiotherapy revealed significant effects with COMT rs4646316, GSTP1 rs1965, and XPC rs2228001. Major sources of variations between studies include differences in patient demographics, ototoxicity grading systems, and treatment protocols. CONCLUSION Our meta-analysis presents polymorphisms that exert ototoxic or otoprotective effects in patients undergoing PBC. Importantly, several of these alleles are observed at high frequencies globally, highlighting the potential for polygenic screening and cumulative risk evaluation for personalized care.
Collapse
Affiliation(s)
- Daniel Z Hong
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Thaned C C Ong
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Dhayan P Timbadia
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Hui T A Tan
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Eunice D Kwa
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Wan Q Chong
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Boon C Goh
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Woei S Loh
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Kwok S Loh
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Ene C Tan
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Joshua K Tay
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| |
Collapse
|
16
|
Freyer DR, Orgel E, Knight K, Krailo M. Special considerations in the design and implementation of pediatric otoprotection trials. J Cancer Surviv 2023; 17:4-16. [PMID: 36637630 DOI: 10.1007/s11764-022-01312-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/07/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE Cisplatin-induced hearing loss (CIHL) is a common late effect after childhood cancer treatment having profound, lifelong consequences that lower quality of life. The recent identification of intravenous sodium thiosulfate (STS) as an effective agent for preventing pediatric CIHL represents a paradigm shift that has created new opportunities for expanding STS usage and developing additional otoprotectants. The purpose of this paper is to discuss key considerations and recommendations for the design and implementation of future pediatric otoprotection trials. METHODS An approach synthesizing published data and collective experience was used. RESULTS Key issues were identified in the categories of translational research, trial designs for systemic and intratympanic agents, measurement of ototoxicity, and biostatistical challenges. CONCLUSIONS Future pediatric otoprotection trials should emphasize (1) deep integration of preclinical and early-phase studies; (2) an embedded or free-standing design for systemic agents based on mechanistic considerations; (3) use of suitable audiologic testing batteries for children, SIOP grading criteria, and submission of raw audiologic data for central review; and (4) novel endpoints and innovative study designs that maximize trial efficiency for limited sample sizes. Additional recommendations include routine collection of DNA specimens for assessing modifying effects of genetic susceptibility and meaningful inclusion of patient/family advocates for informing trial development. IMPLICATIONS FOR CANCER SURVIVORS Changing the historical paradigm from acceptance to prevention of pediatric CIHL through expanded research with existing and emerging otoprotectants will dramatically improve quality of life for future childhood cancer survivors exposed to cisplatin.
Collapse
Affiliation(s)
- David R Freyer
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Departments of Pediatrics, Medicine, and Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Etan Orgel
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kristin Knight
- Department of Audiology, Doernbecher Children's Hospital, Portland, OR, USA
- Oregon Health and Science University, Portland, OR, USA
| | - Mark Krailo
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
YTHDF1 Protects Auditory Hair Cells from Cisplatin-Induced Damage by Activating Autophagy via the Promotion of ATG14 Translation. Mol Neurobiol 2022; 59:7134-7151. [PMID: 36097301 DOI: 10.1007/s12035-022-03021-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/29/2022] [Indexed: 01/05/2023]
Abstract
N6-methyladenosine (m6A) has been recognized as a common type of post-transcriptional epigenetic modification. m6A modification and YTHDF1, one of its reader proteins, have been documented to play a pivotal role in numerous human diseases via regulating mRNA splicing, translation, stability, and subcellular localization. The chemotherapeutic drug cisplatin (CDP) can damage sensory hair cells (HCs) and result in permanent sensorineural hearing loss. However, whether YTHDF1-mediated modification of mRNA is potentially involved in CDP-induced injury in sensory hair cells was not fully clarified. This study investigated the potential mechanisms for the modification of YTHDF1 in CDP-induced damage in HCs. Here, we discovered that YTHDF1's expression level statistically increased significantly after treating with CDP. Apoptosis and cell death of HCs induced by CDP were exacerbated after the knockdown of YTHDF1, while overexpression of YTHDF1 in HCs alleviated their injury induced by CDP. Moreover, YTHDF1 expression correlated with cisplatin-induced autophagy with statistical significance in HCs; namely, YTHDF1's overexpression enhanced the activation of autophagy, while its deficiency suppressed autophagy and, at the same time, increased the loss of HCs after CDP damage. WB analysis and qRT-PCR results of autophagy-related genes indicated that YTHDF1 promoted the translation of autophagy-related genes ATG14, thus boosting autophagy. Therefore, CDP-induced YTHDF1 expression protected HCs against CDP-induced apoptosis by upregulating the translation of autophagy-related genes ATG14, along with enhancing autophagy. Based on these findings, it can be inferred that YTHDF1 is potentially a target for ameliorating drug-induced HCs damage through m6A modification.
Collapse
|
18
|
Attar M, Alqarni MS, Alsinnari YM, Bukhari ZM, Alshegifi H, Alzhrani A, Alshaikh K, Alsubaie H, Muqat M, Alhakami H, Algarni M. The Incidence and Risk Factors of Cisplatin and Carboplatin Ototoxicity in Pediatric Oncology Patients at Tertiary Oncology Center. Indian J Surg Oncol 2022; 13:925-930. [PMID: 36687225 PMCID: PMC9845443 DOI: 10.1007/s13193-022-01579-7] [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: 12/19/2021] [Accepted: 06/28/2022] [Indexed: 01/25/2023] Open
Abstract
Pediatric cancers are relatively rare diseases when considering all types of cancer. Platinum-based chemotherapeutic agents are potent agents against a variety of pediatric malignancies. An important adverse effect of platinum-based agents is the occurrence of hearing loss. This hearing loss can pose a challenge to detect especially if the child is in his early of life. It will also significantly affect the child development of social, pedagogical, and personal dimensions. It is integral to identify incidence of platinum-based ototoxicity and risk factors that increase the likelihood of developing hearing loss in cancer children. We performed a retrospective chart review of 123 pediatric patients who had completed cisplatin and carboplatin therapy for a variety of malignancies. Patients were diagnosed at Princess Nourah Oncology Centre between January 2011 and December 2016, were less than 14 years old at diagnosis. Audiograms were scored using the International Society of Pediatric Oncology (SIOP) Boston Scale (0-4), a validated grading system for cisplatin-related hearing loss. Ototoxicity was reported in 16 patients out of 123 with a rate of 13%. The incidence of ototoxicity was highest in CNS tumors such as medulloblastoma (37.5%) and optic glioma (25%). Males were at greater risk for developing hearing loss than females. Cumulative cisplatin dose and addition radiation therapy were also identified as risk factors for development of ototoxicity (P = 0.008). Nature and location of cancer, gender, cumulative dose, and addition of radiation therapy are important clinical biomarkers of cisplatin ototoxicity.
Collapse
Affiliation(s)
- Meshari Attar
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Mohammed S. Alqarni
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Yaser M. Alsinnari
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Ziad M. Bukhari
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Hussein Alshegifi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Abdulrahman Alzhrani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Khalid Alshaikh
- King Abdulaziz Medical City, National Guard Hospital, Jeddah, Saudi Arabia
| | - Haya Alsubaie
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Abdulaziz Medical City, National Guard Hospital, Jeddah, Saudi Arabia
| | - Mahmoud Muqat
- King Abdulaziz Medical City, National Guard Hospital, Jeddah, Saudi Arabia
| | - Hadi Alhakami
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Abdulaziz Medical City, National Guard Hospital, Jeddah, Saudi Arabia
| | - Mohammed Algarni
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Abdulaziz Medical City, National Guard Hospital, Jeddah, Saudi Arabia
| |
Collapse
|
19
|
Sherief LM, Rifky E, Attia M, Ahmed R, Kamal NM, Oshi MAM, Hanna D. Platinum-induced ototoxicity in pediatric cancer survivors: GSTP1 c.313A>G variant association. Medicine (Baltimore) 2022; 101:e31627. [PMID: 36397425 PMCID: PMC9666226 DOI: 10.1097/md.0000000000031627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hearing damage is one of the main toxic effects of platinum compounds, it derives from the irreversible degeneration of hair cells of the ear. Genetic association studies have suggested an association between GSTP1 c.313A>G variant and platinum-induced ototoxicity in childhood cancer survivors. We aimed to detect the frequency of ototoxicity and associated risk factors in survivors of childhood cancer receiving platinum-based chemotherapy and to detect the relation between GSTP1 c.313A>G (rs1695) polymorphisms and ototoxicity. We conducted a cross-sectional study on 64 cancer survivors who received platinum agents (cisplatin and/or carboplatin) at least 2 years after the end of chemotherapy. The patients underwent comprehensive audiological evaluations and genotyping to detect the presence of the GSTP1 c.313A>G polymorphisms. Hearing loss (HL) was identified in 16/64 patients (25%), including 62.5% treated with cisplatin and 37.5% treated with carboplatin. The greater incidence of ototoxicity was found in children treated for osteosarcoma (28.1%) followed by patients with germ cell tumors (25%) and neuroblastoma (21.9%). The AA, AG, and GG types of GSTP1 c.313A>G variant were detected in 84.4%, 9.4%, and 6.3%, respectively, of patients with HL with a significant association between mutant genotype of GSTP1 rs1695 and platinum-induced ototoxicity (P = .035). HL was not significantly associated with the total cumulative dose of cisplatin and carboplatin. GSTP1 c.313A>G variant may increase the risk of HL in pediatric oncology patients treated with cisplatin or carboplatin chemotherapy.
Collapse
Affiliation(s)
- Laila M. Sherief
- Department of Pediatrics and Pediatric Hematology/Oncology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- * Correspondence: Laila M. Sherief, Department of Pediatrics, Zagazig University, Zagazig, Egypt (e-mail: )
| | - Elhamy Rifky
- Department of Pediatrics and Pediatric Hematology/Oncology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed Attia
- Department of Clinical Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Naglaa M. Kamal
- Department of Pediatrics and Pediatric Hepatology, Faculty of Medicine Cairo University, Cairo, Egypt
| | - Mohammed A. M. Oshi
- Department of Pediatrics and Pediatric Neurology, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Diana Hanna
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
20
|
Romani AM. Cisplatin in Cancer Treatment. Biochem Pharmacol 2022; 206:115323. [DOI: 10.1016/j.bcp.2022.115323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
|
21
|
Strebel S, Mader L, Sláma T, Waespe N, Weiss A, Parfitt R, Am Zehnhoff-Dinnesen A, Kompis M, von der Weid NX, Ansari M, Kuehni CE. Severity of hearing loss after platinum chemotherapy in childhood cancer survivors. Pediatr Blood Cancer 2022; 69:e29755. [PMID: 35723448 DOI: 10.1002/pbc.29755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/25/2022] [Accepted: 04/15/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Hearing loss is a potential side effect from childhood cancer treatment. We described the severity of hearing loss assessed by audiometry in a representative national cohort of childhood cancer survivors (CCS) and identified clinical risk factors. PROCEDURE We included all CCS from the Swiss Childhood Cancer Registry who were diagnosed ≤18 age and treated with platinum-based chemotherapy between 1990 and 2014. We extracted audiograms, treatment-related information, and demographic data from medical records. Two reviewers independently assessed the severity of hearing loss at latest follow-up using the Münster Ototoxicity Scale. We used ordered logistic regression to identify clinical risk factors for severity of hearing loss. RESULTS We analyzed data from 270 CCS. Median time from cancer diagnosis to last audiogram was 5 years (interquartile range 2.5-8.1 years). We found 53 (20%) CCS with mild, 78 (29%) with moderate, and 75 (28%) with severe hearing loss. Higher severity grades were associated with (a) younger age at cancer diagnosis (odds ratio [OR] 5.4, 95% confidence interval [CI]: 2.5-12.0 for <5 years); (b) treatment in earlier years (OR 4.8, 95% CI: 2.1-11.0 for 1990-1995); (c) higher cumulative cisplatin doses (OR 13.5, 95% CI: 4.7-38.8 for >450 mg/m2 ); (d) concomitant cranial radiation therapy (CRT) (OR 4.4, 95% CI: 2.5-7.8); and (e) hematopoietic stem cell transplantation (HSCT) (OR 2.7, 95% CI: 1.0-7.2). CONCLUSION Three of four CCS treated with platinum-based chemotherapy experienced some degree of hearing loss. We recommend closely monitoring patient's hearing function if treated at a young age with high cumulative cisplatin doses, and concomitant CRT as part of long-term care.
Collapse
Affiliation(s)
- Sven Strebel
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,CANSEARCH research platform in pediatric oncology and hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Luzius Mader
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Tomáš Sláma
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Nicolas Waespe
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,CANSEARCH research platform in pediatric oncology and hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Annette Weiss
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Bavarian Care and Nursing Authority, Amberg, Germany
| | - Ross Parfitt
- Department for Phoniatrics and Pedaudiology, University Hospital Münster, University of Münster, Münster, Germany
| | | | - Martin Kompis
- Department of ENT, Head and Neck Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicolas X von der Weid
- Department of Pediatric Oncology and Hematology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Marc Ansari
- CANSEARCH research platform in pediatric oncology and hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Department of Women, Child and Adolescent, Division of Pediatric Oncology and Hematology, Geneva University Hospital, University of Geneva, Geneva, Switzerland
| | - Claudia E Kuehni
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Coffin AB, Dale E, Doppenberg E, Fearington F, Hayward T, Hill J, Molano O. Putative COVID-19 therapies imatinib, lopinavir, ritonavir, and ivermectin cause hair cell damage: A targeted screen in the zebrafish lateral line. Front Cell Neurosci 2022; 16:941031. [PMID: 36090793 PMCID: PMC9448854 DOI: 10.3389/fncel.2022.941031] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
The biomedical community is rapidly developing COVID-19 drugs to bring much-need therapies to market, with over 900 drugs and drug combinations currently in clinical trials. While this pace of drug development is necessary, the risk of producing therapies with significant side-effects is also increased. One likely side-effect of some COVID-19 drugs is hearing loss, yet hearing is not assessed during preclinical development or clinical trials. We used the zebrafish lateral line, an established model for drug-induced sensory hair cell damage, to assess the ototoxic potential of seven drugs in clinical trials for treatment of COVID-19. We found that ivermectin, lopinavir, imatinib, and ritonavir were significantly toxic to lateral line hair cells. By contrast, the approved COVID-19 therapies dexamethasone and remdesivir did not cause damage. We also did not observe damage from the antibiotic azithromycin. Neither lopinavir nor ritonavir altered the number of pre-synaptic ribbons per surviving hair cell, while there was an increase in ribbons following imatinib or ivermectin exposure. Damage from lopinavir, imatinib, and ivermectin was specific to hair cells, with no overall cytotoxicity noted following TUNEL labeling. Ritonavir may be generally cytotoxic, as determined by an increase in the number of TUNEL-positive non-hair cells following ritonavir exposure. Pharmacological inhibition of the mechanotransduction (MET) channel attenuated damage caused by lopinavir and ritonavir but did not alter imatinib or ivermectin toxicity. These results suggest that lopinavir and ritonavir may enter hair cells through the MET channel, similar to known ototoxins such as aminoglycoside antibiotics. Finally, we asked if ivermectin was ototoxic to rats in vivo. While ivermectin is not recommended by the FDA for treating COVID-19, many people have chosen to take ivermectin without a doctor's guidance, often with serious side-effects. Rats received daily subcutaneous injections for 10 days with a clinically relevant ivermectin dose (0.2 mg/kg). In contrast to our zebrafish assays, ivermectin did not cause ototoxicity in rats. Our research suggests that some drugs in clinical trials for COVID-19 may be ototoxic. This work can help identify drugs with the fewest side-effects and determine which therapies warrant audiometric monitoring.
Collapse
Affiliation(s)
- Allison B. Coffin
- Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, United States
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| | - Emily Dale
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| | - Emilee Doppenberg
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| | - Forrest Fearington
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| | - Tamasen Hayward
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| | - Jordan Hill
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| | - Olivia Molano
- College of Arts and Sciences, Washington State University, Vancouver, WA, United States
| |
Collapse
|
23
|
Dillard LK, Lopez-Perez L, Martinez RX, Fullerton AM, Chadha S, McMahon CM. Global burden of ototoxic hearing loss associated with platinum-based cancer treatment: A systematic review and meta-analysis. Cancer Epidemiol 2022; 79:102203. [PMID: 35724557 PMCID: PMC9339659 DOI: 10.1016/j.canep.2022.102203] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 11/06/2022]
Abstract
Platinum-based chemotherapeutic agents cisplatin and carboplatin are widely used in cancer treatment worldwide and may result in ototoxic hearing loss. The high incidence of cancer and salient ototoxic effects of platinum-based compounds pose a global public health threat. The purpose of this study was twofold. First, to estimate the prevalence of ototoxic hearing loss associated with treatment with cisplatin and/or carboplatin via a systematic review and meta-analysis. Second, to estimate the annual global burden of ototoxic hearing loss associated with exposure to cisplatin and/or carboplatin. For the systematic review, three databases were searched (Ovid Medline, Ovid Embase, and Web of Science Core Collection) and studies that reported prevalence of objectively measured ototoxic hearing loss in cancer patients were included. A random effects meta-analysis determined pooled prevalence (95% confidence intervals [CI]) of ototoxic hearing loss overall, and estimates were stratified by treatment and patient attributes. Estimates of ototoxic hearing loss burden were created with published global estimates of incident cancers often treated with platinum-based compounds and cancer-specific treatment rates. Eighty-seven records (n = 5077 individuals) were included in the meta-analysis. Pooled prevalence of ototoxic hearing loss associated with cisplatin and/or carboplatin exposure was 43.17% [CI 37.93-48.56%]. Prevalence estimates were higher for regimens involving cisplatin (cisplatin only: 49.21% [CI 42.62-55.82%]; cisplatin & carboplatin: 56.05% [CI 45.12-66.43%]) versus carboplatin only (13.47% [CI 8.68-20.32%]). Our crude estimates of burden indicated approximately one million individuals worldwide are likely exposed to cisplatin and/or carboplatin, which would result in almost half a million cases of hearing loss per year, globally. There is an urgent need to reduce impacts of ototoxicity in cancer patients. This can be partially achieved by implementing existing strategies focused on primary, secondary, and tertiary hearing loss prevention. Primary ototoxicity prevention via otoprotectants should be a research and policy priority.
Collapse
Affiliation(s)
- Lauren K Dillard
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, United States.
| | - Lucero Lopez-Perez
- Cluster of Healthier Populations, World Health Organization, Geneva, Switzerland
| | - Ricardo X Martinez
- Cluster of Healthier Populations, World Health Organization, Geneva, Switzerland
| | - Amanda M Fullerton
- Department of Linguistics, Macquarie University, Sydney, New South Wales, Australia
| | - Shelly Chadha
- Department on Noncommunicable Diseases, World Health Organization, Geneva, Switzerland
| | - Catherine M McMahon
- Department of Linguistics, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Harrison RT, DeBacker JR, Trevino M, Bielefeld EC, Lobarinas E. Cochlear Preconditioning as a Modulator of Susceptibility to Hearing Loss. Antioxid Redox Signal 2022; 36:1215-1228. [PMID: 34011160 DOI: 10.1089/ars.2021.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance: Acquired sensorineural hearing loss is a major public health problem worldwide. The leading causes of sensorineural hearing loss are noise, aging, and ototoxic medications, with the key underlying pathology being damage to the cochlea. The review focuses on the phenomenon of preconditioning, in which the susceptibility to cochlear injury is reduced by exposing the ear to a stressful stimulus. Recent Advances: Cochlear conditioning has focused on the use of mono-modal conditioning, specifically conditioning the cochlea with moderate noise exposures before a traumatic exposure that causes permanent hearing loss. Recently, cross-modal conditioning has been explored more thoroughly, to prevent not only noise-induced hearing loss, but also age-related and drug-induced hearing losses. Critical Issues: Noise exposures that cause only temporary threshold shifts (TTSs) can cause long-term synaptopathy, injury to the synapses between the inner hair cells and spiral ganglion cells. This discovery has the potential to significantly alter the field of cochlear preconditioning with noise. Further, cochlear preconditioning can be the gateway to the development of clinically deployable therapeutics. Therefore, understanding the underlying mechanisms of conditioning is crucial for optimizing clinical protection against sensorineural hearing loss. Future Directions: Before the discovery of synaptopathy, noise exposures that caused only TTSs were believed to be either harmless or potentially beneficial. Any considerations of preconditioning with noise must consider the potential for injury to the synapses. Further, the discovery of different methods to precondition the cochlea against injury will yield new avenues for protection against hearing loss in the vulnerable populations. Antioxid. Redox Signal. 36, 1215-1228.
Collapse
Affiliation(s)
- Ryan T Harrison
- Department of Speech and Hearing Science, The Ohio State University, Columbus, Ohio, USA
| | - J Riley DeBacker
- Department of Speech and Hearing Science, The Ohio State University, Columbus, Ohio, USA
| | - Monica Trevino
- Callier Center for Communication Disorders, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas, USA
| | - Eric C Bielefeld
- Department of Speech and Hearing Science, The Ohio State University, Columbus, Ohio, USA
| | - Edward Lobarinas
- Callier Center for Communication Disorders, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
25
|
Fetoni AR, Paciello F, Troiani D. Cisplatin Chemotherapy and Cochlear Damage: Otoprotective and Chemosensitization Properties of Polyphenols. Antioxid Redox Signal 2022; 36:1229-1245. [PMID: 34731023 DOI: 10.1089/ars.2021.0183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Significance: Cisplatin is an important component of treatment regimens for different cancers. Notwithstanding that therapeutic success often results from partial efficacy or stabilizing the disease, chemotherapy failure is driven by resistance to drug treatment and occurrence of side effects, such as progressive irreversible ototoxicity. Cisplatin's side effects, including ototoxicity, are often dose limiting. Recent Advances: Cisplatin ototoxicity results from several mechanisms, including redox imbalance caused by reactive oxygen species production and lipid peroxidation, activation of inflammation, and p53 and its downstream pathways that culminate in apoptosis. Considerable efforts in research have targeted development of molecular interventions that can be concurrently administered with cisplatin or other chemotherapies to reduce side effect toxicities while preserving or enhancing the antineoplastic effects. Evidence from studies has indicated some polyphenols, such as curcumin, can help to regulate redox signaling and inflammatory effects. Furthermore, polyphenols can exert opposing effects in different types of tissues, that is, normal cells undergoing stressful conditions versus cancer cells. Critical Issues: This review article summarizes evidence of curcumin antioxidant effect against cisplatin-induced ototoxicity that is converted to a pro-oxidant activity in cisplatin-treated cancer cells, thus providing an ideal chemosensitivity combined with otoprotection. Polyphenols can modulate the adaptive responses to stress in the cisplatin-exposed cochlea. These adaptive effects can result from the interaction/cross talk between the cell's defenses, inflammatory molecules, and the key signaling molecules of signal transducers and activators of transcription 3 (STAT-3), nuclear factor κ-B (NF-κB), p53, and nuclear factor erythroid 2-related factor 2 (Nrf-2). Future Directions: We provide molecular evidence for alternative strategies for chemotherapy with cisplatin addressing the otoprotection and chemosensitization properties of polyphenols. Antioxid. Redox Signal. 36, 1229-1245.
Collapse
Affiliation(s)
- Anna Rita Fetoni
- Department of Head and Neck Surgery, Università Cattolica Del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Troiani
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
26
|
Pharmacological treatment with annexin A1-derived peptide protects against cisplatin-induced hearing loss. Toxicol Lett 2022; 363:27-35. [PMID: 35561849 DOI: 10.1016/j.toxlet.2022.05.001] [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: 11/30/2021] [Revised: 04/11/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022]
Abstract
Cisplatin is an antineoplastic agent widely used, and no effective treatments capable of preventing cisplatin-induced ototoxicity and neurotoxicity in humans have yet been identified. This study evaluated the effect of the anti-inflammatory annexin A1 (AnxA1)-derived peptide Ac2-26 in a cisplatin-induced ototoxicity model. Wistar rats received intraperitoneal injections of cisplatin (10mg/kg/day) for 3 days to induce hearing loss, and Ac2-26 (1mg/kg) was administered 15minutes before cisplatin administration. Control animals received an equal volume of saline. Hearing thresholds were measured by distortion product otoacoustic emissions (DPOAE) before and after treatments. Pharmacological treatment with Ac2-26 protected against cisplatin-induced hearing loss, as evidenced by DPOAE results showing similar signal-noise ratios between the control and Ac2-26-treated groups. These otoprotective effects of Ac2-26 were associated with an increased number of ganglion neurons compared with the untreated cisplatin group. Additionally, Ac2-26 treatment produced reduced immunoreactivity on cleaved caspase 3 and phosphorylated ERK levels in the ganglion neurons, compared to the untreated group, supporting the neuroprotective effects of the Ac2-26. Our results suggest that Ac2-26 has a substantial otoprotective effect in this cisplatin-induced ototoxicity model mediated by neuroprotection and the regulation of the ERK pathway.
Collapse
|
27
|
Medical Therapy of Hearing Impairment and Tinnitus with Chinese Medicine: An Overview. Chin J Integr Med 2022:10.1007/s11655-022-3678-5. [PMID: 35419727 DOI: 10.1007/s11655-022-3678-5] [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: 10/13/2021] [Indexed: 11/03/2022]
Abstract
The current review gives a comprehensive overview of the recent development in Chinese medicine (CM) for treating several kinds of acquired nerve deafness and tinnitus, as well as links the traditional principle to well-established pharmacological mechanisms for future research. To date, about 24 herbal species and 40 related ingredients used in CM to treat hearing loss and tinnitus are reported for the treatment of endocochlear potential, endolymph growth, lowering toxic and provocative substance aggregation, inhibiting sensory cell death, and retaining sensory transfer. However, there are a few herbal species that can be used for medicinal purposes. Nevertheless, clinical studies have been hampered by a limited population sample, a deficiency of a suitable control research group, or contradictory results. Enhanced cochlear blood flow, antiinflammatory antioxidant, neuroprotective effects, and anti-apoptotic, as well as multi-target approach on different auditory sections of the inner ear, are all possible benefits of CM medications. There are numerous unknown natural products for aural ailment and tinnitus identified in CM that are expected to be examined in the future utilizing various aural ailment models and processes.
Collapse
|
28
|
Aldrink JH, Glick RD, Baertschiger RM, Kulaylat AN, Lautz TB, Christison-Lagay E, Grant CN, Tracy E, Dasgupta R, Brown EG, Mattei P, Rothstein DH, Rodeberg DA, Ehrlich PF. Update on pediatric testicular germ cell tumors. J Pediatr Surg 2022; 57:690-699. [PMID: 33975708 DOI: 10.1016/j.jpedsurg.2021.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Testicular germ cell tumors are uncommon tumors that are encountered by pediatric surgeons and urologists and require a knowledge of appropriate contemporary evaluation and surgical and medical management. METHOD A review of the recommended diagnostic evaluation and current surgical and medical management of children and adolescents with testicular germ cell tumors based upon recently completed clinical trials was performed and summarized in this article. RESULTS In this summary of childhood and adolescent testicular germ cell tumors, we review the initial clinical evaluation, surgical and medical management, risk stratification, results from recent prospective cooperative group studies, and clinical outcomes. A summary of recently completed clinical trials by pediatric oncology cooperative groups is provided, and best surgical practices are discussed. CONCLUSIONS Testicular germ cell tumors in children are rare tumors. International collaborations, data-sharing, and enrollment of patients at all stages and risk classifications into active clinical trials will enhance our knowledge of these rare tumors and most importantly improve outcomes of patients with testicular germ cell tumors. LEVEL OF EVIDENCE This is a review article of previously published and referenced level 1 and 2 studies, but also includes expert opinion level 5, represented by the American Pediatric Surgical Association Cancer Committee.
Collapse
Affiliation(s)
- Jennifer H Aldrink
- Department of Surgery, Division of Pediatric Surgery, The Ohio State University College of Medicine, Nationwide Children's Hospital, Columbus, OH 43205, United States.
| | - Richard D Glick
- Division of Pediatric Surgery, Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, New Hyde Park, NY, United States
| | - Reto M Baertschiger
- Division of General and Thoracic Surgery, The Hospital for Sick Kids, University of Toronto, Toronto, Ontario, Canada
| | - Afif N Kulaylat
- Division of Pediatric Surgery, Department of Surgery, Penn State Children's Hospital, Hershey, PA, United States
| | - Timothy B Lautz
- Division of Pediatric Surgery, Department of Surgery, Ann and Robert H Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL, United States
| | - Emily Christison-Lagay
- Department of Surgery, Division of Pediatric Surgery, Yale-New Haven Children's Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Christa N Grant
- Division of Pediatric Surgery, Department of Surgery, Penn State Children's Hospital, Hershey, PA, United States
| | - Elisabeth Tracy
- Department of Surgery, Division of Pediatric Surgery, Duke University Medical Center, Durham, NC, United States
| | - Roshni Dasgupta
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Medical Center, University of Cincinnati, Cincinnati OH, United States
| | - Erin G Brown
- Division of Pediatric Surgery, Department of Surgery, University of California Davis, Sacramento, CA, United States
| | - Peter Mattei
- General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - David H Rothstein
- Department of Surgery, Seattle Children's Hospital, University of Washington, Seattle, WA, United States
| | - David A Rodeberg
- Department of Surgery, Division of Pediatric Surgery, East Carolina University, Greenville, NC, United States
| | - Peter F Ehrlich
- Department of Surgery, Mott Children's Hospital, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
29
|
Moon P, Theruvath J, Chang J, Song Y, Shpanskaya K, Maleki M, Cheng AG, Ahmad IN, Yeom KW. MRI Correlates of Ototoxicity in the Auditory Pathway in Children Treated for Medulloblastoma. Otol Neurotol 2022; 43:e97-e104. [PMID: 34739428 DOI: 10.1097/mao.0000000000003336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To assess diffusion and perfusion changes of the auditory pathway in pediatric medulloblastoma patients exposed to ototoxic therapies. STUDY DESIGN Retrospective cohort study. SETTING A single academic tertiary children's hospital. PATIENTS Twenty pediatric medulloblastoma patients (13 men; mean age 12.0 ± 4.8 yr) treated with platinum-based chemotherapy with or without radiation and 18 age-and-sex matched controls were included. Ototoxicity scores were determined using Chang Ototoxicity Grading Scale. INTERVENTIONS Three Tesla magnetic resonance was used for diffusion tensor and arterial spin labeling perfusion imaging. MAIN OUTCOME MEASURES Quantitative diffusion tensor metrics were extracted from the Heschl's gyrus, auditory radiation, and inferior colliculus. Arterial spin labeling perfusion of the Heschl's gyrus was also examined. RESULTS Nine patients had clinically significant hearing loss, or Chang grades more than or equal to 2a; 11 patients had mild/no hearing loss, or Chang grades less than 2a. The clinically significant hearing loss group showed reduced mean diffusivity in the Heschl's gyrus (p = 0.018) and auditory radiation (p = 0.037), and decreased perfusion in the Heschl's gyrus (p = 0.001). Mild/no hearing loss group showed reduced mean diffusivity (p = 0.036) in Heschl's gyrus only, with a decrease in perfusion (p = 0.008). There were no differences between groups in the inferior colliculus. There was no difference in fractional anisotropy between patients exposed to ototoxic therapies and controls. CONCLUSIONS Patients exposed to ototoxic therapies demonstrated microstructural and physiological alteration of the auditory pathway. The present study shows proof-of-concept use of diffusion tensor imaging to gauge ototoxicity along the auditory pathway. Future larger cohort studies are needed to assess significance of changes in diffusion tensor imaging longitudinally, and the relationship between these changes and hearing loss severity and longitudinal changes of the developing auditory white matter.
Collapse
Affiliation(s)
| | | | | | - Yohan Song
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Katie Shpanskaya
- Department of Radiology, Duke University School of Medicine, Durham, North Carolina
| | - Maryam Maleki
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine
| | - Iram N Ahmad
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine
| | - Kristen W Yeom
- Department of Radiology, Lucile Packard Children's Hospital, Stanford, California
| |
Collapse
|
30
|
Patatt FSA, Gonçalves LF, Paiva KMD, Haas P. Ototoxic effects of antineoplastic drugs: a systematic review. Braz J Otorhinolaryngol 2022; 88:130-140. [PMID: 33757754 PMCID: PMC9422719 DOI: 10.1016/j.bjorl.2021.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/11/2021] [Accepted: 02/06/2021] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Platinum-based chemotherapeutics play an important role in the treatment of cancer at different levels and are the most cited ototoxic agents when scientific evidence is analyzed. OBJECTIVE To present scientific evidence based on a systematic literature review, PRISMA, in order to systematize information on the ototoxic effects of using antineoplastic drugs. METHODS For the selection of studies, the combination based on the Medical Subject Heading Terms (MeSH) was used. The Medline (Pubmed), LILACS, SciELO, SCOPUS, WEB OF SCIENCE and BIREME databases were used, without restriction of language, period, and location. Evaluation of the quality of the articles was carried out, which included articles with a minimum score of 6 in the modified scale of the literature. The designs of the selected studies were descriptive, cohort, and cross-sectional, which were related to the research objective. RESULTS Three articles were included in this systematic review. The ototoxicity caused by cisplatin alone varied from 45% to 83.3%, while that caused by the use associated with carboplatin varied from 16.6% to 75%. There was a significant variation in the cumulative doses of these antineoplastic agents, both in isolated and in combination. Auditory changes, especially at high frequencies, were evident after completion of treatment. CONCLUSION Auditory changes after the use of platinum-based antineoplastic drugs were found, however, there was an important heterogeneity regarding the frequency of ototoxicity and the cumulative dose of the drugs used.
Collapse
Affiliation(s)
| | | | | | - Patrícia Haas
- Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| |
Collapse
|
31
|
Sánchez-Canteli M, Núñez-Batalla F, Martínez-González P, de Lucio-Delgado A, Antonio Villegas-Rubio J, Gómez-Martínez JR, Luis Llorente-Pendás J. Ototoxicity in cancer survivors: Experience and proposal of a surveillance protocol. An Pediatr (Barc) 2021; 95:290-297. [PMID: 34702687 DOI: 10.1016/j.anpede.2020.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/26/2020] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Ototoxicity occurs in different percentages in patients after treatment with platinum-based chemotherapy or cranial radiation therapy. The aim of this study was to present our experience in ototoxicity monitoring. MATERIAL AND METHODS A review was made of the registry of paediatric cancer patients referred to the Children's Hearing Loss Unit from 1999 to 2019. RESULTS Of the 46 patients referred to this unit, 41 had received platinum as part of their treatment, 17 patients underwent neurosurgery, and 18 patients received cranial radiation therapy. An anamnesis and otoscopy were performed on all of them, and the monitoring was carried out with tone-verbal audiometry and/or distortion products. Hearing loss was observed in eight patients (21.05% of patients referred for audiological follow-up) as a consequence of the treatment. It was impossible to determine the audiological situation in eight patients at the end of treatment. Hearing aid adaption was necessary in two patients. In coordination with Paediatric Oncology, a change from cisplatin to carboplatin due to bilateral grade two ototoxicity was considered appropriate during treatment in one patient. CONCLUSION Adequate coordination with Paediatric Oncology is essential to carry out active surveillance for ototoxicity and to modify, if possible, the dosage or type of chemotherapy in case hearing is affected. In our experience, and following current recommendations, a pre-treatment assessment is usually performed, as well as monitoring during treatment, at the end of treatment, and annually thereafter due to the risk of a later development of hearing loss.
Collapse
Affiliation(s)
- Mario Sánchez-Canteli
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.
| | - Faustino Núñez-Batalla
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Universidad de Oviedo, Oviedo, Spain
| | - Patricia Martínez-González
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana de Lucio-Delgado
- Oncología Pediátrica, Servicio de Pediatría, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Justo Ramón Gómez-Martínez
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Universidad de Oviedo, Oviedo, Spain
| | - José Luis Llorente-Pendás
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
32
|
Campbell KC, Rehemtulla A, Sunkara P, Hamstra D, Buhnerkempe M, Ross B. Oral D-methionine protects against cisplatin-induced hearing loss in humans: phase 2 randomized clinical trial in India. Int J Audiol 2021; 61:621-631. [PMID: 34622731 DOI: 10.1080/14992027.2021.1983215] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objective: This exploratory Phase 2 clinical trial is the first determining safety and efficacy of oral D-methionine (D-met) in reducing cisplatin-induced ototoxicity.Design: Randomised parallel double-blind placebo-controlled exploratory Phase 2 study.Study samples: Fifty adult cancer patients received oral D-met or placebo before each cisplatin dose. Physical examination, blood collection and audiometry occurred at baseline and subsequent visits plus post-treatment audiometry. After attrition, final analysis included 27 patients.Results: Significant treatment group by ear and time (baseline vs. post-treatment) interactions occurred at 10 kHz and 11.2 kHz. Placebo and D-met groups differed in threshold shift for left ear at 11.2 kHz (mean difference = 22.97 dB [9.59, 36.35]). Averaging across ears, placebo group showed significant threshold shifts from baseline to post-treatment at 10 kHz (mean shift= -13.65 dB [-21.32,-5.98]), 11.2 kHz (-16.15 dB [-25.19,-7.12]), and 12.5 kHz (-11.46 dB [-19.18,-3.74]) but not 8 kHz (-8.65 dB [-17.86, 0.55]). The D-met group showed no significant threshold shifts (8 kHz: -1.25 dB [-7.75, 5.25]; 10 kHz:-3.93 dB [-8.89, 1.03]; 11.2 kHz:-4.82 dB [-11.21, 1.57]; 12.5 kHz:-3.68 dB [-11.57, 4.21]). Side effects did not significantly differ between groups.Conclusion: Oral D-met reduces cisplatin-induced ototoxicity in humans.
Collapse
Affiliation(s)
- Kathleen C Campbell
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Alnawez Rehemtulla
- Molecular Therapeutics, Molecular Cancer Therapeutics, Ann Arbor, MI, USA
| | | | - Daniel Hamstra
- Department of Radiation Oncology, William Beaumont Oakland University Medical School, Dearborn, MI, USA
| | - Michael Buhnerkempe
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Brian Ross
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Olgun Y, Çakir Kizmazoğlu D, İnce D, Ellidokuz H, Güneri EA, Olgun N, Kirkim G. Evaluation of Risk Factors Causing Ototoxicity in Childhood Cancers Located in the Head and Neck Region Treated With Platinum-based Chemotherapy. J Pediatr Hematol Oncol 2021; 43:e930-e934. [PMID: 33885038 DOI: 10.1097/mph.0000000000002158] [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: 08/08/2020] [Accepted: 02/22/2021] [Indexed: 10/21/2022]
Abstract
The aim of this study is to evaluate risk factors contributing to the development of ototoxicity in children who received platinum-based chemotherapy for malignancies located in the head and neck region. Eighty-four children who received platinum-based chemotherapy were included. Audiologic evaluations were performed before and after each chemotherapy session through pure tone audiometry, distortion product otoacoustic emissions, and auditory brainstem response tests. Ototoxicity was evaluated using Brock, Muenster, and Chang classifications. Factors such as cranial irradiation, cumulative doses of cisplatin, age, sex, cotreatment with aminoglycosides, schedule of platinum, and type of chemotherapeutic agent were analyzed. Using χ2 tests, all risk factors were matched with the 3 ototoxicity classifications, and multivariate analyses were conducted using statistically significant risk factors. In univariate analyses, being between 5 and 12 years of age, cranial irradiation and being treated with both cisplatin and carboplatin were found to be related to ototoxicity in all 3 classifications. Logistic regression modeling analyses with these 3 risk factors showed that being between 5 and 12 years of age and being treated with both cisplatin and carboplatin significantly increased the risk of ototoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Günay Kirkim
- HearingSpeech and Balance Unit, Dokuz Eylül University School of Medicine, Izmir, Turkey
| |
Collapse
|
34
|
Minturn JE, Mochizuki AY, Partap S, Belasco JB, Lange BJ, Li Y, Phillips PC, Gibbs IC, Fisher PG, Fisher MJ, Janss AJ. A Pilot Study of Low-Dose Craniospinal Irradiation in Patients With Newly Diagnosed Average-Risk Medulloblastoma. Front Oncol 2021; 11:744739. [PMID: 34540703 PMCID: PMC8443797 DOI: 10.3389/fonc.2021.744739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/17/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Medulloblastoma is one of the most common malignant brain tumors in children. To date, the treatment of average-risk (non-metastatic, completely resected) medulloblastoma includes craniospinal radiation therapy and adjuvant chemotherapy. Modern treatment modalities and now risk stratification of subgroups have extended the survival of these patients, exposing the long-term morbidities associated with radiation therapy. Prior to advances in molecular subgrouping, we sought to reduce the late effects of radiation in patients with average-risk medulloblastoma. Methods We performed a single-arm, multi-institution study, reducing the dose of craniospinal irradiation by 25% to 18 Gray (Gy) with the goal of maintaining the therapeutic efficacy as described in CCG 9892 with maintenance chemotherapy. Results Twenty-eight (28) patients aged 3-30 years were enrolled across three institutions between April 2001 and December 2010. Median age at enrollment was 9 years with a median follow-up time of 11.7 years. The 3-year relapse-free (RFS) and overall survival (OS) were 79% (95% confidence interval [CI] 58% to 90%) and 93% (95% CI 74% to 98%), respectively. The 5-year RFS and OS were 71% (95% CI 50% to 85%) and 86% (95% CI 66% to 94%), respectively. Toxicities were similar to those seen in other studies; there were no grade 5 toxicities. Conclusions Given the known neurocognitive adverse effects associated with cranial radiation therapy, studies to evaluate the feasibility of dose reduction are needed. In this study, we demonstrate that select patients with average-risk medulloblastoma may benefit from a reduced craniospinal radiation dose of 18 Gy without impacting relapse-free or overall survival. Clinical Trial Registration ClinicalTrials.gov identifier: NCT00031590
Collapse
Affiliation(s)
- Jane E Minturn
- Department of Pediatrics, Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Aaron Y Mochizuki
- Department of Pediatrics, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Neurology and Neurological Sciences, Division of Child Neurology, Lucile Packard Children's Hospital at Stanford University, Palo Alto, CA, United States
| | - Sonia Partap
- Department of Neurology and Neurological Sciences, Division of Child Neurology, Lucile Packard Children's Hospital at Stanford University, Palo Alto, CA, United States
| | - Jean B Belasco
- Department of Pediatrics, Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Beverly J Lange
- Department of Pediatrics, Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Yimei Li
- Department of Pediatrics, Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Peter C Phillips
- Department of Pediatrics, Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Iris C Gibbs
- Department of Neurology and Neurological Sciences, Division of Child Neurology, Lucile Packard Children's Hospital at Stanford University, Palo Alto, CA, United States.,Department of Radiation Oncology, Stanford University Cancer Center, Palo Alto, CA, United States
| | - Paul G Fisher
- Department of Neurology and Neurological Sciences, Division of Child Neurology, Lucile Packard Children's Hospital at Stanford University, Palo Alto, CA, United States
| | - Michael J Fisher
- Department of Pediatrics, Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Anna J Janss
- Department of Pediatrics, Division of Hematology/Oncology, Children's Healthcare of Atlanta, Atlanta, GA, United States
| |
Collapse
|
35
|
Meijer AJM, Li KH, Brooks B, Clemens E, Ross CJ, Rassekh SR, Hoetink AE, van Grotel M, van den Heuvel-Eibrink MM, Carleton BC. The cumulative incidence of cisplatin-induced hearing loss in young children is higher and develops at an early stage during therapy compared with older children based on 2052 audiological assessments. Cancer 2021; 128:169-179. [PMID: 34490624 DOI: 10.1002/cncr.33848] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/17/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Ototoxicity is a common adverse event of cisplatin treatment. The authors investigated the development of cisplatin-induced hearing loss (CIHL) over time in children with cancer by age and examined the influence of other clinical characteristics on the course of CIHL. METHODS Data from Canadian patients with childhood cancer were retrospectively reviewed. Hearing loss was graded according to International Society of Pediatric Oncology criteria. The Kaplan-Meier method was applied to estimate the cumulative incidence of CIHL for the total cohort and according to age. Cox regression models were used to explore the effects of independent variables on CIHL development up to 3 years after the start of therapy. RESULTS In total, 368 patients with 2052 audiological assessments were included. Three years after initiating therapy, the cumulative incidence of CIHL was highest in patients aged ≤5 years (75%; 95% confidence interval [CI], 66%-84%), with a rapid increase observed to 27% (95% CI, 21%-35%) at 3 months and to 61% (95% CI, 53%-69%) at 1 year, compared with patients aged >5 years (48%; 95% CI, 37%-62%; P < .001). The total cumulative dose of cisplatin at 3 months (per 100 mg/m2 increase: hazard ratio [HR], 1.20; 95% CI, 1.01-1.41) vincristine (HR, 2.87; 95% CI, 1.89-4.36) and the total duration of concomitantly administered antibiotics (>30 days: HR, 1.85; 95% CI, 1.17-2.95) further influenced CIHL development over time. CONCLUSIONS In young children, the cumulative incidence of CIHL is higher compared with that in older children and develops early during therapy. The course of CIHL is further influenced by the total cumulative dose of cisplatin and other ototoxic (co-)medication. These results highlight the need for audiological monitoring at each cisplatin cycle.
Collapse
Affiliation(s)
- Annelot J M Meijer
- Department of Pediatric Oncology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kathy H Li
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Beth Brooks
- Department of Audiology and Speech Pathology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada.,School of Audiology and Speech Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eva Clemens
- Department of Pediatric Oncology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Colin J Ross
- Pharmaceutical Outcomes Program, British Columbia Children's Hospital, Vancouver, British Columbia, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sharad R Rassekh
- Division of Hematology, Oncology, and Bone Marrow Transplant, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alex E Hoetink
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Utrecht-Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Martine van Grotel
- Department of Pediatric Oncology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Pediatric Oncology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Bruce C Carleton
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Program, British Columbia Children's Hospital, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
36
|
Meijer AJM, van den Heuvel-Eibrink MM, Brooks B, Am Zehnhoff-Dinnesen AG, Knight KR, Freyer DR, Chang KW, Hero B, Papadakis V, Frazier AL, Blattmann C, Windsor R, Morland B, Bouffet E, Rutkowski S, Tytgat GAM, Geller JI, Hunter LL, Sung L, Calaminus G, Carleton BC, Helleman HW, Foster JH, Kruger M, Cohn RJ, Landier W, van Grotel M, Brock PR, Hoetink AE, Rajput KM. Recommendations for Age-Appropriate Testing, Timing, and Frequency of Audiologic Monitoring During Childhood Cancer Treatment: An International Society of Paediatric Oncology Supportive Care Consensus Report. JAMA Oncol 2021; 7:1550-1558. [PMID: 34383016 DOI: 10.1001/jamaoncol.2021.2697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Ototoxicity is an irreversible direct and late effect of certain childhood cancer treatments. Audiologic surveillance during therapy as part of the supportive care pathway enables early detection of hearing loss, decision-making about ongoing cancer treatment, and, when applicable, the timely use of audiologic interventions. Pediatric oncologic clinical practice and treatment trials have tended to be driven by tumor type and tumor-specific working groups. Internationally accepted standardized recommendations for monitoring hearing during treatment have not previously been agreed on. Objective To provide standard recommendations on hearing loss monitoring during childhood cancer therapy for clinical practice. Methods An Ototoxicity Task Force was formed under the umbrella of the International Society of Paediatric Oncology, consisting of international audiologists, otolaryngologists, and leaders in the field of relevant pediatric oncology tumor groups. Consensus meetings conducted by experts were organized, aimed at providing standardized recommendations on age-directed testing, timing, and frequency of monitoring during cancer treatment based on literature and consensus. Consensus statements were prepared by the core group, adapted following several videoconferences, and finally agreed on by the expert panel. Findings The consensus reached was that children who receive ototoxic cancer treatment (platinum agents, cranial irradiation, and/or brain surgery) require a baseline case history, monitoring of their middle ear and inner ear function, and assessment of tinnitus at each audiologic follow-up. As a minimum, age-appropriate testing should be performed before and at the end of treatment. Ideally, audiometry with counseling before each cisplatin cycle should be considered in the context of the individual patient, specific disease, feasibility, and available resources. Conclusions and Relevance This is an international multidisciplinary consensus report providing standardized supportive care recommendations on hearing monitoring in children undergoing potentially ototoxic cancer treatment. The recommendations are intended to improve the care of children with cancer and facilitate comparative research on the timing and development of hearing loss caused by different cancer treatment regimens.
Collapse
Affiliation(s)
- Annelot J M Meijer
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Beth Brooks
- Department of Audiology and Speech Pathology, BC Children's Hospital, Vancouver, British Columbia, Canada.,School of Audiology and Speech Science, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Kristin R Knight
- Department of Pediatric Audiology, Oregon Health and Science University, Portland
| | - David R Freyer
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles.,Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles.,Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles
| | - Kay W Chang
- Department of Otolaryngology, Stanford University School of Medicine, Palo Alto, California
| | - Barbara Hero
- Department of Pediatric Hematology-Oncology, Children's Hospital, University of Cologne, Cologne, Germany
| | - Vassilios Papadakis
- Department of Pediatric Hematology-Oncology, Agia Sofia Children's Hospital, Athens, Greece
| | - A Lindsay Frazier
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Claudia Blattmann
- Department of Pediatric Oncology/Hematology/Immunology, Stuttgart Cancer Center, Olgahospital Stuttgart, Stuttgart, Germany
| | - Rachael Windsor
- Department of Oncology, University College London Hospitals National Health Service Trust, London, United Kingdom
| | - Bruce Morland
- Department of Pediatric Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Hamburg, Germany
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Lisa L Hunter
- Communication Sciences Research Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Lillian Sung
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Child Health Evaluative Sciences, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Canada
| | - Gabriele Calaminus
- Department of Pediatric Hematology and Oncology, University Hospital Bonn, Bonn, Germany
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Hiske W Helleman
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Utrecht-Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Jennifer H Foster
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Houston, Texas
| | - Mariana Kruger
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Richard J Cohn
- Kids Cancer Centre, Sydney Children's Hospital, High Street, Randwick, Australia.,School of Women's and Children's Health, University of New South Wales Medicine, Sydney, Australia
| | - Wendy Landier
- Department of Pediatrics, Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham
| | - Martine van Grotel
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Penelope R Brock
- Department of Pediatric Oncology, Great Ormond Street Hospital for Children National Health Service Trust, London, United Kingdom
| | - Alexander E Hoetink
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Utrecht-Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Kaukab M Rajput
- Department of Audiovestibular Medicine and Cochlear Implant, Great Ormond Street Hospital for Children National Health Service Trust, London, United Kingdom
| | | |
Collapse
|
37
|
Sex Differences in the Triad of Acquired Sensorineural Hearing Loss. Int J Mol Sci 2021; 22:ijms22158111. [PMID: 34360877 PMCID: PMC8348369 DOI: 10.3390/ijms22158111] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022] Open
Abstract
The triad of noise-generated, drug-induced, and age-related hearing loss is the major cause of acquired sensorineural hearing loss (ASNHL) in modern society. Although these three forms of hearing loss display similar underlying mechanisms, detailed studies have revealed the presence of sex differences in the auditory system both in human and animal models of ASNHL. However, the sexual dimorphism of hearing varies among noise-induced hearing loss (NIHL), ototoxicity, and age-related hearing loss (ARHL). Importantly, estrogen may play an essential role in modulating the pathophysiological mechanisms in the cochlea and several reports have shown that the effects of hormone replacement therapy on hearing loss are complex. This review will summarize the clinical features of sex differences in ASNHL, compare the animal investigations of cochlear sexual dimorphism in response to the three insults, and address how estrogen affects the auditory organ at molecular levels.
Collapse
|
38
|
Meijer AJM, Diepstraten FA, Langer T, Broer L, Domingo IK, Clemens E, Uitterlinden AG, de Vries ACH, van Grotel M, Vermeij WP, Ozinga RA, Binder H, Byrne J, van Dulmen-den Broeder E, Garrè ML, Grabow D, Kaatsch P, Kaiser M, Kenborg L, Winther JF, Rechnitzer C, Hasle H, Kepak T, Kepakova K, Tissing WJE, van der Kooi ALF, Kremer LCM, Kruseova J, Pluijm SMF, Kuehni CE, van der Pal HJH, Parfitt R, Spix C, Tillmanns A, Deuster D, Matulat P, Calaminus G, Hoetink AE, Elsner S, Gebauer J, Haupt R, Lackner H, Blattmann C, Neggers SJCMM, Rassekh SR, Wright GEB, Brooks B, Nagtegaal AP, Drögemöller BI, Ross CJD, Bhavsar AP, Am Zehnhoff-Dinnesen AG, Carleton BC, Zolk O, van den Heuvel-Eibrink MM. TCERG1L allelic variation is associated with cisplatin-induced hearing loss in childhood cancer, a PanCareLIFE study. NPJ Precis Oncol 2021; 5:64. [PMID: 34262104 PMCID: PMC8280110 DOI: 10.1038/s41698-021-00178-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/16/2021] [Indexed: 12/24/2022] Open
Abstract
In children with cancer, the heterogeneity in ototoxicity occurrence after similar treatment suggests a role for genetic susceptibility. Using a genome-wide association study (GWAS) approach, we identified a genetic variant in TCERG1L (rs893507) to be associated with hearing loss in 390 non-cranial irradiated, cisplatin-treated children with cancer. These results were replicated in two independent, similarly treated cohorts (n = 192 and 188, respectively) (combined cohort: P = 5.3 × 10-10, OR 3.11, 95% CI 2.2-4.5). Modulating TCERG1L expression in cultured human cells revealed significantly altered cellular responses to cisplatin-induced cytokine secretion and toxicity. These results contribute to insights into the genetic and pathophysiological basis of cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- A J M Meijer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | - F A Diepstraten
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - T Langer
- Department of Pediatric Oncology and Hematology, University Hospital for Children and Adolescents, Lübeck, Germany
| | - L Broer
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - I K Domingo
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - E Clemens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - A G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - M van Grotel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - W P Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - R A Ozinga
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - H Binder
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - J Byrne
- Boyne Research Institute, Drogheda, Ireland
| | - E van Dulmen-den Broeder
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- VU Medical Center, Amsterdam, The Netherlands
| | - M L Garrè
- Department of Neurooncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - D Grabow
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - P Kaatsch
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - M Kaiser
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - L Kenborg
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - J F Winther
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University and University Hospital, Aarhus, Denmark
| | - C Rechnitzer
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - H Hasle
- Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | - T Kepak
- University Hospital Brno, Brno, Czech Republic
- International Clinical Research Center (FNUSA-ICRC), Brno, Czech Republic
| | - K Kepakova
- University Hospital Brno, Brno, Czech Republic
- International Clinical Research Center (FNUSA-ICRC), Brno, Czech Republic
| | - W J E Tissing
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A L F van der Kooi
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Obstetrics and Gynecology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - L C M Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - J Kruseova
- Department of Children Hemato-Oncology, Motol University Hospital Prague, Prague, Czech Republic
| | - S M F Pluijm
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - C E Kuehni
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Pediatric Hematology and Oncology, University Children's Hospital Bern, University of Bern, Bern, Switzerland
| | - H J H van der Pal
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - R Parfitt
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - C Spix
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - A Tillmanns
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - D Deuster
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - P Matulat
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - G Calaminus
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - A E Hoetink
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Utrecht, Utrecht, The Netherlands
| | - S Elsner
- Institute of Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | - J Gebauer
- Department of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - R Haupt
- Epidemiology and Biostatistics Unit and DOPO Clinic, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - H Lackner
- Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - C Blattmann
- Department of Pediatric Oncology/Hematology/Immunology, Stuttgart Cancer Center, Olgahospital, Stuttgart, Germany
| | - S J C M M Neggers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - S R Rassekh
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - G E B Wright
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - B Brooks
- Audiology and Speech Pathology Department, BC Children's Hospital, Vancouver, BC, Canada
| | - A P Nagtegaal
- Departement of Otorhinolaryngology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - B I Drögemöller
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, British Columbia, Canada
| | - C J D Ross
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, British Columbia, Canada
| | - A P Bhavsar
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - A G Am Zehnhoff-Dinnesen
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - B C Carleton
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, British Columbia, Canada
| | - O Zolk
- Institute of Clinical Pharmacology, Brandenburg Medical School, Rüdersdorf, Germany
| | - M M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Pro S, Vinti L, Boni A, Mastronuzzi A, Scilipoti M, Velardi M, Caroleo AM, Farina E, Badolato F, Alessi I, Di Nardo G, Carai A, Valeriani M, Reale A, Parisi P, Raucci U. Peripheral Nervous System Involvement in Non-Primary Pediatric Cancer: From Neurotoxicity to Possible Etiologies. J Clin Med 2021; 10:3016. [PMID: 34300182 PMCID: PMC8303855 DOI: 10.3390/jcm10143016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 01/21/2023] Open
Abstract
Peripheral neuropathy is a well described complication in children with cancer. Oncologists are generally well aware of the toxicity of the main agents, but fear the side effects of new drugs. As chemotherapeutic agents have been correlated with the activation of the immune system such as in Chemotherapy Induced Peripheral Neuropathy (CIPN), an abnormal response can lead to Autoimmune Peripheral Neuropathy (APN). Although less frequent but more severe, Radiation Induced Peripheral Neuropathy may be related to irreversible peripheral nervous system (PNS). Pediatric cancer patients also have a higher risk of entering a Pediatric Intensive Care Unit for complications related to therapy and disease. Injury to peripheral nerves is cumulative, and frequently, the additional stress of a malignancy and its therapy can unmask a subclinical neuropathy. Emerging risk factors for CIPN include treatment factors such as dose, duration and concurrent medication along with patient factors, namely age and inherited susceptibilities. The recent identification of individual genetic variations has advanced the understanding of physiopathological mechanisms and may direct future treatment approaches. More research is needed on pharmacological agents for the prevention or treatment of the condition as well as rehabilitation interventions, in order to allow for the simultaneous delivery of optimal cancer therapy and the mitigation of toxicity associated with pain and functional impairment. The aim of this paper is to review literature data regarding PNS complications in non-primary pediatric cancer.
Collapse
Affiliation(s)
- Stefano Pro
- Child Neurology Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.P.); (M.V.)
| | - Luciana Vinti
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.V.); (A.M.); (A.M.C.); (I.A.)
| | - Alessandra Boni
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.B.); (E.F.)
| | - Angela Mastronuzzi
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.V.); (A.M.); (A.M.C.); (I.A.)
| | - Martina Scilipoti
- Department of Emergency, Acceptance and General Pediatrics, Bambino Gesù Children Hospital, IRCCS, 00165 Rome, Italy; (M.S.); (A.R.)
| | - Margherita Velardi
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (M.V.); (F.B.); (G.D.N.); (P.P.)
| | - Anna Maria Caroleo
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.V.); (A.M.); (A.M.C.); (I.A.)
| | - Elisa Farina
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (A.B.); (E.F.)
| | - Fausto Badolato
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (M.V.); (F.B.); (G.D.N.); (P.P.)
| | - Iside Alessi
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.V.); (A.M.); (A.M.C.); (I.A.)
| | - Giovanni Di Nardo
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (M.V.); (F.B.); (G.D.N.); (P.P.)
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Massimiliano Valeriani
- Child Neurology Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.P.); (M.V.)
| | - Antonino Reale
- Department of Emergency, Acceptance and General Pediatrics, Bambino Gesù Children Hospital, IRCCS, 00165 Rome, Italy; (M.S.); (A.R.)
| | - Pasquale Parisi
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy; (M.V.); (F.B.); (G.D.N.); (P.P.)
| | - Umberto Raucci
- Department of Emergency, Acceptance and General Pediatrics, Bambino Gesù Children Hospital, IRCCS, 00165 Rome, Italy; (M.S.); (A.R.)
| |
Collapse
|
40
|
Camet ML, Spence A, Hayashi SS, Wu N, Henry J, Sauerburger K, Hayashi RJ. Cisplatin Ototoxicity: Examination of the Impact of Dosing, Infusion Times, and Schedules In Pediatric Cancer Patients. Front Oncol 2021; 11:673080. [PMID: 34262862 PMCID: PMC8274419 DOI: 10.3389/fonc.2021.673080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Background Sensorineural hearing loss is a well-known side effect of cisplatin (CDDP). There is limited research on the effect of dosing, infusion times, and schedules of cisplatin administration and their impact on hearing loss. Methods A retrospective review of 993 pediatric patients' medical and audiological charts from August 1990 to March 2015 was conducted using stringent inclusion criteria to characterize patients with hearing loss. 248 of these patients received CDDP. Of these, 216 patients had sufficient CDDP infusion data to assess for sensorineural hearing loss attributable to CDDP and its associated risk factors. Chart reviews were performed to extract clinical data including CDDP dosing information. Demographic and clinical characteristics were summarized by descriptive statistics, and univariate and multivariate logistic regressions were performed to examine the relationship between hearing loss and specific parameters of cisplatin administration (amount infused per dose, prescribed infusion time, total number of doses, number of doses per cycle, number of cycles, cumulative cisplatin exposure). Stepwise variable selection procedure was performed in the multivariate model building to extract the best subset of risk factors for the prediction of hearing loss and worsening ototoxicity grade using an established ototoxicity grading scale from the International Society of Pediatric Oncology (SIOP). Results A total of 153 patients with complete medical and audiologic data were evaluable for analysis. Hearing loss was identified in 72.6% of the patients. Multivariate analysis revealed that age [OR=0.90 (0.84-0.97), p-value=0.0086], radiation to any part of the body, [OR=3.20 (1.29-7.93), p-value=0.012], amount infused per dose (mg/m2) [OR=1.018 (1.002-1.033), p-value=0.029], and cumulative cisplatin exposure (mg/m 2) [OR=1.004 (1-1.008), p-value=0.027] were associated with hearing loss. Similar associations were also found between these risk factors and worsening SIOP grade. Conclusion In one of the largest studies examining the influence of CDDP dosing and schedules on hearing loss, we found the amount of CDDP infused per dose is a significant risk factor. Considerations in designing regimens that reduce the amount of CDDP infused per dose may reduce the risk of hearing loss. Randomized prospective trials are needed.
Collapse
Affiliation(s)
- Miranda L Camet
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Anne Spence
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Susan S Hayashi
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ningying Wu
- Biostatistics Shared Resource, Division of Public Health Sciences, Department of Surgery, The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Jennifer Henry
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kara Sauerburger
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Robert J Hayashi
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO, United States
| |
Collapse
|
41
|
Mohri H, Ninoyu Y, Sakaguchi H, Hirano S, Saito N, Ueyama T. Nox3-Derived Superoxide in Cochleae Induces Sensorineural Hearing Loss. J Neurosci 2021; 41:4716-4731. [PMID: 33849947 PMCID: PMC8260246 DOI: 10.1523/jneurosci.2672-20.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 01/02/2023] Open
Abstract
Reactive oxygen species (ROS) produced by NADPH oxidases (Nox) contribute to the development of different types of sensorineural hearing loss (SNHL), a common impairment in humans with no established treatment. Although the essential role of Nox3 in otoconia biosynthesis and its possible involvement in hearing have been reported in rodents, immunohistological methods targeted at detecting Nox3 expression in inner ear cells reveal ambiguous results. Therefore, the mechanism underlying Nox3-dependent SNHL remains unclear and warrants further investigation. We generated Nox3-Cre knock-in mice, in which Nox3 was replaced with Cre recombinase (Cre). Using Nox3-Cre;tdTomato mice of either sex, in which tdTomato is expressed under the control of the Nox3 promoter, we determined Nox3-expressing regions and cell types in the inner ear. Nox3-expressing cells in the cochlea included various types of supporting cells, outer hair cells, inner hair cells, and spiral ganglion neurons. Nox3 expression increased with cisplatin, age, and noise insults. Moreover, increased Nox3 expression in supporting cells and outer hair cells, especially at the basal turn of the cochlea, played essential roles in ROS-related SNHL. The extent of Nox3 involvement in SNHL follows the following order: cisplatin-induced hearing loss > age-related hearing loss > noise-induced hearing loss. Here, on the basis of Nox3-Cre;tdTomato, which can be used as a reporter system (Nox3-Cre+/-;tdTomato+/+ and Nox3-Cre+/+;tdTomato+/+), and Nox3-KO (Nox3-Cre+/+;tdTomato+/+) mice, we demonstrate that Nox3 inhibition in the cochlea is a promising strategy for ROS-related SNHL, such as cisplatin-induced HL, age-related HL, and noise-induced HL.SIGNIFICANCE STATEMENT We found Nox3-expressing regions and cell types in the inner ear, especially in the cochlea, using Nox3-Cre;tdTomato mice, a reporter system generated in this study. Nox3 expression increased with cisplatin, age, and noise insults in specific cell types in the cochlea and resulted in the loss (apoptosis) of outer hair cells. Thus, Nox3 might serve as a molecular target for the development of therapeutics for sensorineural hearing loss, particularly cisplatin-induced, age-related, and noise-induced hearing loss.
Collapse
Affiliation(s)
- Hiroaki Mohri
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yuzuru Ninoyu
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| | - Hirofumi Sakaguchi
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Shigeru Hirano
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Naoaki Saito
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| | - Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| |
Collapse
|
42
|
Issa AM, Aboushawareb SA, Eisenstat DD, Guilcher GM, Liu G, Rassekh SR, Strahlendorf C, Tallen G, Tanoshima R, Carleton B. Deliberations about clinical pharmacogenetic testing in pediatric oncology. Per Med 2021; 18:399-405. [PMID: 33973801 DOI: 10.2217/pme-2020-0120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
This article summarizes the background, content and outcomes of a special meeting that was convened among oncologists and scientists to discuss the role of pharmacogenetic (PGx) testing in pediatric clinical oncology practice. This meeting provided an opportunity for what the lead author (AM Issa) refers to as the 'voice of the clinician' dynamic to be amplified in order to better understand how personalized or precision medicine applications such as PGx testing are adopted and incorporated into clinical settings and what we can learn from the experiences of current and ongoing implementation PGx approaches to further the implementation of precision medicine applications in real-world environments. Group dynamics and clinical experience with PGx testing and return of results shaped the discussion.
Collapse
Affiliation(s)
- Amalia M Issa
- Personalized Precision Medicine & Targeted Therapeutics Institute, PA 19064, USA.,Departments of Pharmaceutical Sciences and Health Policy, University of The Sciences in Philadelphia, Philadelphia, PA 19104, USA.,Centre of Genomics & Policy, McGill University, Montreal, Quebec H3A 0G1, Canada.,Department of Family Medicine, McGill University, Montreal, Quebec H3S 1Z1, Canada
| | | | - David D Eisenstat
- Department of Paediatrics, Murdoch Children's Research Institute, University of Melbourne, Melbourne 3052, Australia.,Departments of Pediatrics, Medical Genetics & Oncology, University of Alberta, Edmonton AB T6G 2H7, Canada
| | - Greg Mt Guilcher
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta T3B 6A8, Canada.,Department of Oncology, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Geoffrey Liu
- Department of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto ON M5G 2C1, Canada
| | - S Rod Rassekh
- Department of Pediatrics & BC Children's Hospital Research Institute, Division of Translational Therapeutics, University of British Columbia, Vancouver, BC V6H 3V4, Canada.,YCU Center for Novel & Exploratory Clinical Trials, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan.,Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| | - Caron Strahlendorf
- Department of Pediatrics & BC Children's Hospital Research Institute, Division of Translational Therapeutics, University of British Columbia, Vancouver, BC V6H 3V4, Canada.,YCU Center for Novel & Exploratory Clinical Trials, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan.,Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| | - Gesche Tallen
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta T3B 6A8, Canada.,Pediatric Hematology/Oncology/BMT, BC Children's Hospital, Vancouver BC V6H 3V4, Canada
| | - Reo Tanoshima
- Department of Pediatric Oncology/Hematology, Charité-Medical School Charitéplatz 1, Berlin 10117, Germany.,Department of Pediatrics, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
| | - Bruce Carleton
- Department of Pediatrics & BC Children's Hospital Research Institute, Division of Translational Therapeutics, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| |
Collapse
|
43
|
Babolmorad G, Latif A, Domingo IK, Pollock NM, Delyea C, Rieger AM, Allison WT, Bhavsar AP. Toll-like receptor 4 is activated by platinum and contributes to cisplatin-induced ototoxicity. EMBO Rep 2021; 22:e51280. [PMID: 33733573 PMCID: PMC8097357 DOI: 10.15252/embr.202051280] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptor 4 (TLR4) recognizes bacterial lipopolysaccharide (LPS) and can also be activated by some Group 9/10 transition metals, which is believed to mediate immune hypersensitivity reactions. In this work, we test whether TLR4 can be activated by the Group 10 metal platinum and the platinum-based chemotherapeutic cisplatin. Cisplatin is invaluable in childhood cancer treatment but its use is limited due to a permanent hearing loss (cisplatin-induced ototoxicity, CIO) adverse effect. We demonstrate that platinum and cisplatin activate pathways downstream of TLR4 to a similar extent as the known TLR4 agonists LPS and nickel. We further show that TLR4 is required for cisplatin-induced inflammatory, oxidative, and cell death responses in hair cells in vitro and for hair cell damage in vivo. Finally, we identify a TLR4 small molecule inhibitor able to curtail cisplatin toxicity in vitro. Thus, our findings indicate that TLR4 is a promising therapeutic target to mitigate CIO.
Collapse
Affiliation(s)
- Ghazal Babolmorad
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Asna Latif
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Ivan K Domingo
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Niall M Pollock
- Department of Biological SciencesFaculty of ScienceUniversity of AlbertaEdmontonABCanada
| | - Cole Delyea
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Aja M Rieger
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - W Ted Allison
- Department of Biological SciencesFaculty of ScienceUniversity of AlbertaEdmontonABCanada
- Department of Medical GeneticsFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Amit P Bhavsar
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
- Department of Medical GeneticsFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
44
|
Long-term follow-up of high-risk neuroblastoma survivors treated with high-dose chemotherapy and stem cell transplantation rescue. Bone Marrow Transplant 2021; 56:1984-1997. [PMID: 33824435 DOI: 10.1038/s41409-021-01258-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/08/2021] [Accepted: 02/23/2021] [Indexed: 11/08/2022]
Abstract
Intensive treatments including high-dose chemotherapy (HDC) with autologous stem cell rescue have improved high-risk neuroblastoma (HRNB) survival. We report the long-term health status of 145 HRNB survivors, alive and disease-free 5 years post HDC. Median follow-up was 15 years (range = 5-34). Six patients experienced late relapses, 11 developed second malignant neoplasms (SMNs), and 9 died. Event-free and overall survivals 20 years post HDC were 82% (95% CI = 70%-90%) and 89% (78%-95%), respectively. Compared with the French general population, the standardized mortality ratio was 19 (95% CI = 8.7-36.1; p < 0.0001) and the absolute excess risk was 37.6 (19.2-73.5). Late effects were observed in 135/145 patients (median = 3 events/patient); 103 had at least one severe event. SMNs arose at a median of 20 years post HDC and included carcinoma (n = 5), sarcoma (2), acute myeloid leukemia (2), melanoma (1), and malignant glioma (1). Non-oncologic health events included dental maldevelopment (60%), severe hearing loss (20% cumulative probability at 15 years), hepatic focal nodular hyperplasia (14%), thyroid (11%), cardiac (8%), and renal (7%) diseases and growth retardation (height-for-age z-score ≤ -2 for 21%). Gonadal insufficiency was near-universal after busulfan (40/43 females, 33/35 males). Severe late effects are frequent and progressive in HRNB survivors needing systematic very long-term follow-up.
Collapse
|
45
|
Stanford JK, Morgan DS, Bosworth NA, Proctor G, Chen T, Palmer TT, Thapa P, Walters BJ, Vetter DE, Black RD, Rogers LL, Spankovich C. Cool OtOprotective Ear Lumen (COOL) Therapy for Cisplatin-induced Hearing Loss. Otol Neurotol 2021; 42:466-474. [PMID: 33351563 PMCID: PMC8207522 DOI: 10.1097/mao.0000000000002948] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
HYPOTHESIS Localized cooling of the external ear has a protective effect on the susceptibility to cisplatin-induced hearing loss. BACKGROUND We previously demonstrated significant protection from cisplatin-induced hearing loss using cool water ear canal irrigation. However, the study was limited to a single bolus injection of cisplatin and an acute time period. Here, we examined the application of localized cooling of the ear canal with repeated doses of cisplatin, over an expanded period of time, and using two methods of cooling. METHODS Twenty-four guinea pigs (12 male and 12 female) underwent auditory physiological testing (auditory brainstem response and distortion product otoacoustic emissions at 8-32 kHz) and pre/postadministration of cisplatin. Cisplatin (4 mg/kg i.p.) was administered in 3 weekly single injections for a total of 12 mg/kg. While anesthetized, the left ears of the guinea pigs were exposed to either cool water (22°C; ICS Water Caloric Irrigator), a cool ear bar (15°C, cooled by a Peltier device; TNM, Scion NeuroStim), or left uncooled as a sham control. The animals were tested 3 days post each dosage and 1 month post the final dose. At the end of the experiment the animals were euthanized for histological evaluation. RESULTS We found that hearing loss was significantly reduced, and hair cell survival greatly improved, in animals that received cooling treatments compared to cisplatin-only control animals. No significant difference was observed between the two methods of cooling. CONCLUSION Localized cooling of the ear canal during administration of cisplatin mitigated loss of auditory function and loss of hair cells.
Collapse
Affiliation(s)
| | | | | | | | - Tianwen Chen
- Department of Otolaryngology-Head and Neck Surgery
| | | | - Punam Thapa
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bradley J. Walters
- Department of Otolaryngology-Head and Neck Surgery
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Douglas E. Vetter
- Department of Otolaryngology-Head and Neck Surgery
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | | | | | | |
Collapse
|
46
|
An audit of UK audiological practice in specialist paediatric oncology centres regarding hearing assessment of children at risk of ototoxicity due to chemotherapy. The Journal of Laryngology & Otology 2021; 135:14-20. [PMID: 33487183 DOI: 10.1017/s0022215121000025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Platinum-based chemotherapy drugs are associated with substantial ototoxicity. The hearing of children treated with these drugs should be closely monitored. METHOD A questionnaire was sent out to the 19 audiology departments associated with national paediatric cancer specialist centres in the UK looking at current practice in ototoxicity monitoring. RESULTS Responses were received from 17 of 19 centres (89 per cent). All offered some form of audiometric monitoring service. Extended high-frequency testing (9-20 kHz) was only utilised by 7 services (29 per cent). A majority of respondents were reluctant to consider self-test devices in paediatric ototoxicity monitoring (n = 9; 53 per cent). Provision of long-term audiological follow up is sporadic with only 4 (23 per cent) respondents keeping all children with normal hearing under review once treatment is completed. CONCLUSION While some good practice in paediatric ototoxicity was identified, opportunities exist to improve clinical practice and protocols, promote multidisciplinary team working and to utilise technologies such as extended high frequency and self-test audiometry.
Collapse
|
47
|
Hoyer M, Goli R, Barnett KK, Raabe EH, Hong K. Treatment of Hepatoblastoma With Drug-eluting Bead Transarterial Chemoembolization in a 13-Month-Old Infant: A Case Report and Review of the Literature. J Pediatr Hematol Oncol 2021; 43:e123-e126. [PMID: 32459718 DOI: 10.1097/mph.0000000000001842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Prior reports have shown the utility of conventional lipiodol-based transarterial chemoembolization in hepatoblastoma. The authors describe the first reported case of hepatoblastoma treated with drug-eluting bead transarterial chemoembolization (DEB-TACE). OBSERVATIONS An 11-month-old infant presented with hepatoblastoma measuring 14.3 cm. A trial of cisplatin chemotherapy followed by sequential DEB-TACE to the tumor's feeding vasculature reduced the mass to 5.3 cm. The patient tolerated both sessions of DEB-TACE without any major complication. Having demonstrated adequate disease control, the patient then underwent successful liver transplantation. CONCLUSION This report provides promising evidence for the treatment of hepatoblastoma with DEB-TACE.
Collapse
Affiliation(s)
- Matthew Hoyer
- Department of Radiology and Radiological Science, Division of Interventional Radiology
| | - Rakesh Goli
- Department of Radiology and Radiological Science, Division of Interventional Radiology
| | - Katherine K Barnett
- Department of Oncology, Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eric H Raabe
- Department of Oncology, Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kelvin Hong
- Department of Radiology and Radiological Science, Division of Interventional Radiology
| |
Collapse
|
48
|
Landier W, Cohn RJ, van den Heuvel-Eibrink MM. Hearing and Other Neurologic Problems. Pediatr Clin North Am 2020; 67:1219-1235. [PMID: 33131543 DOI: 10.1016/j.pcl.2020.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Ototoxicity and other neurologic toxicities are potential consequences of exposure to common therapeutic agents used during treatment of childhood cancer, including platinum and vinca alkaloid chemotherapy, cranial radiation, surgery involving structures critical to cochlear and neurologic function, and supportive care medications such as aminoglycoside antibiotics and loop diuretics. This article provides an overview of ototoxicity and other neurologic toxicities related to childhood cancer treatment, discusses the challenges that these toxicities may pose for survivors, and presents an overview of current recommendations for surveillance and clinical management of these potentially life-altering toxicities in survivors of childhood cancers.
Collapse
Affiliation(s)
- Wendy Landier
- Pediatric Hematology/Oncology, Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, 1600 7th Avenue South, Lowder 500, Birmingham, AL 35233, USA.
| | - Richard J Cohn
- School of Women's and Children's Health, UNSW Sydney, Medicine, Clinical Oncology, Kids Cancer Centre, Sydney Children's Hospital, High Street, Randwick, Sydney, New South Wales 2031, Australia
| | - Marry M van den Heuvel-Eibrink
- University of Utrecht, Princess Maxima Center for Pediatric Oncology, Prinses Maxima Centrum voor kinderoncologie, Postbus 113 - 3720 AC Bilthoven Heidelberglaan 25, 3584 CS Utrecht, Room number: 2-5 F3, The Netherlands
| |
Collapse
|
49
|
Ototoxicity-induced hearing loss and quality of life in survivors of paediatric cancer. Int J Pediatr Otorhinolaryngol 2020; 138:110401. [PMID: 33152988 DOI: 10.1016/j.ijporl.2020.110401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Hearing loss is a permanent and debilitating side-effect of a range of interventions commonly used in the treatment of childhood cancers, primarily ototoxic medications such as cisplatin. The purpose of this study was to examine the impact of hearing loss, in a population already at risk of reduced quality of life due to the cancer and treatment-related factors. METHODS This study used a questionnaire specifically designed to tap issues relevant to children with hearing loss, the Paediatric Audiology Quality of Life questionnaire (PAQL). Parents of 78 children treated for a wide range of solid tumours and leukaemias completed the PAQL, 41 of whom had sensorineural hearing loss as a result of the cancer treatment. RESULTS Significant differences between those children with hearing loss and those whose hearing remained normal were found on all four scales of the questionnaire. Children affected by ototoxicity were rated as having poorer quality of life in terms of their ability to communicate with family and peers, their independence, interactions with peers and emotional well-being. CONCLUSION These results highlight the impact of acquired hearing loss and reinforce the importance of assessing quality of life with a measure capable of tapping the issues of consequence to the population under investigation. They also have implications for the clinical management of children during and following treatment for a wide range of cancers: medically, audiologically, psychologically and educationally.
Collapse
|
50
|
Sánchez-Canteli M, Núñez-Batalla F, Martínez-González P, de Lucio-Delgado A, Villegas-Rubio JA, Gómez-Martínez JR, Llorente-Pendás JL. [Ototoxicity in cancer survivors: experience and proposal of a surveillance protocol]. An Pediatr (Barc) 2020; 95:S1695-4033(20)30296-4. [PMID: 32998843 DOI: 10.1016/j.anpedi.2020.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Ototoxicity occurs in different percentages in patients after treatment with platinum-based chemotherapy or cranial radiation therapy. The aim of this study was to present experience in ototoxicity monitoring. MATERIAL AND METHODS A review was made of the registry of paediatric cancer patients referred to the Children's Hearing Loss Unit from 1999 to 2019. RESULTS Of the 46 patients referred to this unit, 41 had received platinum as part of their treatment, 17 patients underwent neurosurgery, and 18 patients received cranial radiation therapy. An anamnesis and otoscopy were performed on all of them, and the monitoring was carried out with tone-verbal audiometry and/or distortion products. Hearing loss was observed in eight patients (21.05% of patients referred for audiological follow-up) as a consequence of the treatment. It was impossible to determine the audiological situation in eight patients at the end of treatment. Hearing aid adaption was necessary in two patients. In coordination with Paediatric Oncology, a change from cisplatin to carboplatin due to bilateral grade two ototoxicity was considered appropriate during treatment in one patient. CONCLUSION Adequate coordination with Paediatric Oncology is essential to carry out active surveillance for ototoxicity and to modify, if possible, the dosage or type of chemotherapy in case hearing is affected. In our experience, and following current recommendations, a pre-treatment assessment is usually performed, as well as monitoring during treatment, at the end of treatment, and annually thereafter due to the risk of a later development of hearing loss.
Collapse
Affiliation(s)
- Mario Sánchez-Canteli
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, España; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, España.
| | - Faustino Núñez-Batalla
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, España; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, España; Universidad de Oviedo, Oviedo, España
| | - Patricia Martínez-González
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, España
| | - Ana de Lucio-Delgado
- Oncología Pediátrica, Servicio de Pediatría, Hospital Universitario Central de Asturias, Oviedo, España
| | | | - Justo Ramón Gómez-Martínez
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, España; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, España; Universidad de Oviedo, Oviedo, España
| | - José Luis Llorente-Pendás
- Unidad de Hipoacusia Infantil, Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, España; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, España; Universidad de Oviedo, Oviedo, España
| |
Collapse
|