1
|
Nader ME, Choi J, Hernandez M, Hutcheson K, Myers T, Jivani S, Pratihar R, Fernandez K, Phan J, You C, Gidley PW. The Effects of Metformin on Cisplatin-Induced Ototoxicity in Diabetic Patients. Otolaryngol Head Neck Surg 2025; 172:243-253. [PMID: 39460639 DOI: 10.1002/ohn.1032] [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: 04/19/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVE Ototoxicity is an important side effect of cisplatin. Recent animal and in vitro studies suggest metformin may protect hearing, though human studies are lacking. We report the first retrospective clinical study exploring the effects of metformin on cisplatin-induced ototoxicity. STUDY DESIGN Retrospective case-control. SETTING Tertiary-care center. METHODS We reviewed all diabetic patients treated with cisplatin who were enrolled in an auditory monitoring program between January 1, 2000 and December 31, 2018. Patients were grouped according to their use of metformin at the time of chemotherapy. Demographics, oncologic diagnosis and treatment, and prior noise exposure were recorded. Audiometric thresholds were compared before and after chemotherapy. The primary outcome measure was a change in hearing as defined by the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE). Secondary measures of ototoxicity included changes in hearing using the American Speech-Language-Hearing Association (ASHA) and TUNE criteria. RESULTS Seventy-two diabetic patients met the inclusion criteria. Forty-six patients had taken metformin while on chemotherapy and 26 had not. The proportion of hearing loss as defined by both, CTCAE and ASHA criteria, was more than double in the metformin group relative to the nonmetformin group (CTCAE: 54.4% vs 19.2%, P = .004; ASHA: 62.2% vs 28.0%, P = .003). None of the potential covariates believed to be confounders were significantly associated with the outcome of multivariable analysis. CONCLUSION Contrary to expectations from preclinical data, metformin did not reduce the incidence of hearing loss in patients receiving cisplatin and may, in fact, be associated with an increased risk.
Collapse
Affiliation(s)
- Marc-Elie Nader
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mike Hernandez
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Katherine Hutcheson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Shirin Jivani
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rajarshi Pratihar
- Department of Otorhinolaryngology-Head and Neck Surgery, McGovern Medical School, UTHealth Houston, Houston, Texas, USA
| | - Katharine Fernandez
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Jack Phan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chelsea You
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paul W Gidley
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Fleihan T, Nader ME, Dickman JD. Cisplatin vestibulotoxicity: a current review. Front Surg 2024; 11:1437468. [PMID: 39421409 PMCID: PMC11484025 DOI: 10.3389/fsurg.2024.1437468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Cisplatin, a commonly used chemotherapy drug, is well-established for its ototoxic effects, primarily attributed to the damage it inflicts on cochlear hair cells. However, its impact on the vestibular system remains inadequately understood. Here, we provide a comprehensive review of existing literature concerning cisplatin-induced vestibulotoxicity. Animal studies have shown that cisplatin induces a vestibular hair cell loss that is dose-dependent, with the severity of damage also varying according to the route of administration. Notably, intratympanic and systemic injections in animal models have manifested significant damage primarily to utricular hair cells, with a lesser degree of damage observed for the other vestibular end organs. The underlying mechanisms of cisplatin induced vestibular hair cell loss include apoptosis, oxidative stress, and inflammatory cytokines. Several protective agents, such as Pifithrin-α, DAPT, Ginkgolide B, and heat shock proteins, have demonstrated efficacy in inhibiting cisplatin-induced vestibular damage in preclinical studies. Human clinical findings indicate that cisplatin treatment can cause vestibular dysfunction, characterized by symptoms ranging from transient dizziness to persistent vertigo. Challenges in diagnosis, including the limited utilization of comprehensive vestibular testing for many patients, contribute to the variability in reported outcomes. Cisplatin-induced vestibulotoxicity is a significant complication of chemotherapy, necessitating further research to understand its mechanisms and to improve diagnosis and management, ultimately aiming to enhance the quality of life for cancer patients undergoing cisplatin therapy.
Collapse
Affiliation(s)
- Tamara Fleihan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Marc Elie Nader
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - J. David Dickman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
3
|
Ho JCS, Ma BBY, Chow JCH. Optimizing Hearing Outcomes in Nasopharyngeal Cancer Survivors in the Era of Modern Radiotherapy and Systemic Therapy. Cancers (Basel) 2024; 16:3237. [PMID: 39335208 PMCID: PMC11430699 DOI: 10.3390/cancers16183237] [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: 08/27/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Intensity-modulated radiation therapy (IMRT) improves disease control and reduces treatment-related toxicity in patients with localized nasopharyngeal carcinoma (NPC). However, due to the proximity of the auditory apparatus to the treatment volume and the frequent incorporation of cisplatin-based chemotherapy, treatment-related sensorineural hearing loss (SNHL) remains a common debilitating complication among NPC survivors. The reported crude incidence of SNHL following IMRT for NPC varies widely at 1-46% due to differences in auditory assessment methods and thresholds, follow-up durations, chemotherapy usage, and patient compositions. International guidelines and radiation dosimetric studies have recommended constraining the cochlear mean dose to less than 44-50 Gy, but the risk of SNHL remains high despite adherence to these constraints. Potential strategies to improve hearing outcomes in NPC survivors include cautious de-escalation of radiotherapy dose and volume, individualization of cochlear constraints, optimization of radiotherapy planning techniques, and the use of substitutes or alternative schedules for cisplatin-based chemotherapy. The addition of immune checkpoint inhibitors to chemoradiotherapy did not impact ototoxicity. Prospective studies that employ both objective and patient-reported auditory outcomes are warranted to test the long-term benefits of various approaches. This article aims to provide a comprehensive review of the incidence and radiation dose-toxicity relationship of SNHL in NPC survivors and to summarize potential strategies to optimize hearing outcomes in relation to nuances in radiotherapy planning and the selection of systemic therapy.
Collapse
Affiliation(s)
- Jason C S Ho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Brigette B Y Ma
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - James C H Chow
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| |
Collapse
|
4
|
Duinkerken CW, Chiodo S, Hueniken K, Hauptmann M, Jóźwiak K, Cheng D, Hope A, Liu G, Zuur CL. The role of genetic variants in the prediction of hearing loss due to cisplatin chemoradiotherapy. Cancer Med 2024; 13:e7465. [PMID: 39159054 PMCID: PMC11332395 DOI: 10.1002/cam4.7465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Concomitant high-dose cisplatin with radiotherapy is commonly used for treating head and neck squamous cell carcinoma (HNSCC). Cisplatin, often used with radiotherapy, is known for causing irreversible sensorineural hearing loss, with individual variability suggesting a genetic component. This study aims to enhance the predictive ability of the clinical prediction model for cisplatin-induced hearing loss (CIHL) in HNSCC patients, as outlined in Theunissen et al., by incorporating significant genetic variants. METHODS Conducted at the Netherlands Cancer Institute, this retrospective study included 74 patients treated between 1997 and 2011. Thirty-one SNPs that were previously associated with CIHL or other cisplatin-induced toxicities were identified and incorporated into the model. The primary outcome measured was the change in decibels at posttreatment 1-2-4 kHz hearing levels per additional minor allele of these SNPs, evaluated using linear mixed-effects regression models. The model's predictive accuracy was determined by the area under the curve (AUC) using 10-fold cross-validation. RESULTS The rs2289669 SNP in the SLC47A1/MATE1 gene was linked to a significant 2.67 dB increase in hearing loss per allele (95% CI 0.49-4.86, p = 0.017). Incorporating rs2289669 improved the model's AUC from 0.78 to 0.83, a borderline significant improvement (p = 0.073). CONCLUSIONS This study underscores the importance of the rs2289669 SNP in CIHL and demonstrates the potential of combining genetic and clinical data for enhanced predictive models in personalized treatment strategies.
Collapse
Affiliation(s)
- Charlotte W. Duinkerken
- Department of Otolaryngology and Head and Neck SurgeryLeiden University Medical CentreLeidenthe Netherlands
- Department of Head and Neck Surgerythe Netherlands Cancer InstituteAmsterdamthe Netherlands
| | - Sabrina Chiodo
- Dalla Lana School of Public HealthUniversity of TorontoTorontoCanada
- Department of Medical Oncology and HematologyPrincess Margaret Cancer CentreTorontoCanada
| | - Katrina Hueniken
- Department of BiostatisticsUniversity Health NetworkTorontoCanada
| | - Michael Hauptmann
- Institute of Biostatistics and Registry ResearchBrandenburg Medical School Theodor FontaneNeuruppinGermany
| | - Katarzyna Jóźwiak
- Institute of Biostatistics and Registry ResearchBrandenburg Medical School Theodor FontaneNeuruppinGermany
| | - Dangxiao Cheng
- Department of Medical Oncology and HematologyPrincess Margaret Cancer CentreTorontoCanada
| | - Andrew Hope
- Department of Medical Oncology and HematologyPrincess Margaret Cancer CentreTorontoCanada
| | - Geoffrey Liu
- Dalla Lana School of Public HealthUniversity of TorontoTorontoCanada
- Department of Medical Oncology and HematologyPrincess Margaret Cancer CentreTorontoCanada
| | - Charlotte L. Zuur
- Department of Otolaryngology and Head and Neck SurgeryLeiden University Medical CentreLeidenthe Netherlands
- Department of Head and Neck Surgerythe Netherlands Cancer InstituteAmsterdamthe Netherlands
| |
Collapse
|
5
|
Marin JJG, Serrano MA, Herraez E, Lozano E, Ortiz-Rivero S, Perez-Silva L, Reviejo M, Briz O. Impact of genetic variants in the solute carrier ( SLC) genes encoding drug uptake transporters on the response to anticancer chemotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:27. [PMID: 39143954 PMCID: PMC11322974 DOI: 10.20517/cdr.2024.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 08/16/2024]
Abstract
Cancer drug resistance constitutes a severe limitation for the satisfactory outcome of these patients. This is a complex problem due to the co-existence in cancer cells of multiple and synergistic mechanisms of chemoresistance (MOC). These mechanisms are accounted for by the expression of a set of genes included in the so-called resistome, whose effectiveness often leads to a lack of response to pharmacological treatment. Additionally, genetic variants affecting these genes further increase the complexity of the question. This review focuses on a set of genes encoding members of the transportome involved in drug uptake, which have been classified into the MOC-1A subgroup of the resistome. These proteins belong to the solute carrier (SLC) superfamily. More precisely, we have considered here several members of families SLC2, SLC7, SLC19, SLC22, SLCO, SLC28, SLC29, SLC31, SLC46, and SLC47 due to the impact of their expression and genetic variants in anticancer drug uptake by tumor cells or, in some cases, general bioavailability. Changes in their expression levels and the appearance of genetic variants can contribute to the Darwinian selection of more resistant clones and, hence, to the development of a more malignant phenotype. Accordingly, to address this issue in future personalized medicine, it is necessary to characterize both changes in resistome genes that can affect their function. It is also essential to consider the time-dependent dimension of these features, as the genetic expression and the appearance of genetic variants can change during tumor progression and in response to treatment.
Collapse
Affiliation(s)
- Jose J. G. Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Maria A. Serrano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Elisa Herraez
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Elisa Lozano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Sara Ortiz-Rivero
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Laura Perez-Silva
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Maria Reviejo
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| |
Collapse
|
6
|
Silver JA, Bouganim N, Richardson K, Henry M, Mascarella MA, Ramirez-GarciaLuna J, Golabi N, Mlynarek AM, Zeitouni A, Hier MP, Caglar D, Esfahani K, Sadeghi N. Quality of Life After Neoadjuvant Chemotherapy and Transoral Robotic Surgery for Oropharynx Cancer. JAMA Otolaryngol Head Neck Surg 2024; 150:65-74. [PMID: 38060238 PMCID: PMC10704343 DOI: 10.1001/jamaoto.2023.3781] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/09/2023] [Indexed: 12/08/2023]
Abstract
Importance Efforts are underway to deintensified treatment protocols for patients with human papillomavirus virus-associated oropharyngeal squamous cell carcinoma (HPV-OPSCC) to achieve similar excellent oncologic outcomes while reducing treatment-related adverse effects. Transoral robotic surgery (TORS) as primary treatment often requires adjuvant therapy due to the high incidence of nodal metastasis. Treatment with neoadjuvant chemotherapy followed by TORS and neck dissection (NECTORS), reserving radiation therapy for salvage, yields excellent oncologic outcomes. Objective To assess patient-reported quality of life (QOL) and functional outcomes among patients with HPV-OPSCC who undergo NECTORS. Design, Settings, and Participants This was a multicenter prospective cohort study of patients with HPV-OPSCC treated with the NECTORS protocol in 2017 to 2022. Consecutive patients with stage III or IVa HPV-OPSCC treated with NECTORS in 2017 to 2022 who had completed the primary QOL questionnaire at baseline and at least once during the 24-month follow-up period were included. Ninety-four patients were eligible, and 67 were included in the analyses. Outcome Measures QOL questionnaires at baseline, and at month 1, 3, 6, 12, 18, and 24 posttreatment. Global score on the 30-item European Organization for Research and Treatment of Cancer Core quality of life questionnaire (EORTC QLQ-C30) was the primary outcome; the head and neck extension module (EORTC QLQ-HN35); the MD Anderson Dysphagia Inventory for dysphagia-related QOL; and the Decision Regret Scale were also used. Paired t tests assessed change between the baseline and 12- or 24-month patient-reported outcomes. Results Among the study population of 67 patients (median [range] age, 63 [58-67] years; 54 [80.6%] male) with HPV-OPSCC, the most frequent cancer subsites were palatine tonsil (41 [61%]) and base of tongue (26 [39%]); none required adjuvant RT. Global QOL at 24 months improved compared with baseline (mean difference, 9.49; 95% CI, 2.45 to 16.53). All EORTC QLQ-C30 functional scores returned to baseline or improved within 3 to 6 months posttreatment and remained stable at 24 months. EORTC QLQ-HN35 symptom scale scores improved or were stable at 24 months. The MD Anderson Dysphagia Inventory scores demonstrated no significant difference between baseline and month 12 for global scores (mean difference, 6.15; 95% CI, -4.18 to 16.49) and composite scores (mean difference, 2.73; 95% CI, -1.62 to 7.09). Median (range) score on the Decision Regret Scale was 5 of 100 (0-30), representing mild overall regret. Conclusion and Relevance The findings of this multicenter cohort study indicate that use of the NECTORS protocol is associated with excellent QOL outcomes. QOL measures returned to baseline levels or were better than baseline, which represents positive outcomes for patients with HPV-OPSCC who undergo this treatment regimen.
Collapse
Affiliation(s)
- Jennifer A. Silver
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Nathaniel Bouganim
- Department of Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Keith Richardson
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Melissa Henry
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Oncology, McGill University Health Centre, Montreal, Quebec, Canada
- Lady Davis Research Institute, McGill University, Montreal, Quebec, Canada
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| | - Marco A. Mascarella
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Otolaryngology−Head and Neck Surgery, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | - Nahid Golabi
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alex M. Mlynarek
- Department of Otolaryngology−Head and Neck Surgery, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Anthony Zeitouni
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Michael P. Hier
- Department of Otolaryngology−Head and Neck Surgery, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Derin Caglar
- Department of Pathology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Khashayar Esfahani
- Department of Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Nader Sadeghi
- Department of Otolaryngology−Head and Neck Surgery, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Oncology, McGill University Health Centre, Montreal, Quebec, Canada
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Lee JJW, Alamleh S, Zhan LJ, Hueniken K, Mahler MB, Billfalk-Kelly A, Davies J, Brown MC, Spreafico A, Huang SH, Hope A, Xu W, Goldstein DP, Liu G. Clinical Predictors of Cisplatin Chemoradiation-Induced Ototoxicity in HPV-Positive Oropharyngeal Squamous Cell Carcinoma: A Case-Control Study. J Otolaryngol Head Neck Surg 2024; 53:19160216241248671. [PMID: 39056507 PMCID: PMC11282567 DOI: 10.1177/19160216241248671] [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: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Cisplatin-based chemoradiation is a standard treatment for many patients with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC), an etiologically distinct subset of head and neck cancer. Although associated with good long-term survival, clinical risk factors for ototoxicity have been understudied in this population. This study aimed to evaluate clinical predictors associated with ototoxicity in HPV-positive OPSCC patients treated with cisplatin chemoradiation. METHODS This retrospective case-control study included 201 adult patients (>18 years) with histologically confirmed HPV-positive OPSCC who received cisplatin chemoradiation as their primary treatment from 2001 and 2019 at a single tertiary cancer center. Ototoxicity was determined using baseline and follow-up audiometry and the Common Terminology Criteria for Adverse Events v5.0 grading criteria (Grade ≥2). Multivariable logistic regression [adjusted odds ratio (aOR)] identified significant predictors that increased the odds of ototoxicity. RESULTS A total of 201 patients [165 males; median (IQR) age, 57 (11) years] were included in the study. The incidence of ototoxicity in the worst ear was 56.2%, with the greatest hearing loss occurring at high frequencies (4-8 kHz), resulting in a loss of 12.5 dB at 4 to 6 kHz and 20 dB at 6 to 8 kHz. High-dose cisplatin administration compared to weekly administration [aOR 4.93 (95% CI: 1.84-14.99), P = .003], a higher mean cochlear radiation dose [aOR 1.58 (95% CI: 1.12-2.30), P = .01], smoking history [aOR 2.89 (95% CI: 1.51-5.63), P = .001], and a 10 year increase in age [aOR 2.07 (95% CI: 1.25-3.52), P = .006] were each independently associated with increased odds of ototoxicity. CONCLUSIONS Clinical predictors of ototoxicity in HPV-positive OPSCC patients treated with cisplatin-based chemoradiation include the use of a high-dose cisplatin regimen, higher cochlear radiation doses, a history of smoking, and older age. With the rising incidence of this malignancy in Western countries and overall improved survivorship, our research motivates future studies into risk stratification and earlier interventions to mitigate and reduce the risk of ototoxicity.
Collapse
Affiliation(s)
- John JW. Lee
- Department of Otolaryngology—Head and Neck Surgery, University of Toronto, Toronto, ON, Canada
| | | | - Luna Jia Zhan
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Katrina Hueniken
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Mary B. Mahler
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Astrid Billfalk-Kelly
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Joel Davies
- Department of Otolaryngology—Head and Neck Surgery, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - M. Catherine Brown
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Shao Hui Huang
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Andrew Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - David P. Goldstein
- Department of Otolaryngology—Head and Neck Surgery, University of Toronto, Toronto, ON, Canada
- Department of Otolaryngology—Head and Neck Surgery, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Geoffrey Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
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
|
Han AY, Nader ME, Lam K, Su SY. Current status of sinonasal cancer survivorship care. Head Neck 2023; 45:2458-2468. [PMID: 37449544 DOI: 10.1002/hed.27457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/16/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023] Open
Abstract
Sinonasal cancer is a heterogeneous orphan disease of diverse histologies, each with distinct clinical, oncologic, and toxicity profiles. Because of the comparative rarity of these cancers, sinonasal cancers are treated as a grouped diagnosis despite their clinical and biological heterogeneity. Multimodality treatment with a combination of surgery, chemotherapy, and/or radiotherapy is the standard-of-care for advanced-stage patients but there are few surveillance or follow-up practice guidelines or formalized survivorship care pathways. A scoping literature review was conducted via PubMed, EMBASE, and Google Scholar. A total of 112 studies were included, which were grouped along the following topics: surveillance, second primary tumors, quality of life, and symptom burden. Sinonasal cancer tends to exhibit a higher rate of local failure and occur in a delayed fashion compared to mucosal malignancies of the head and neck. Moreover, the site of failure and time-varying risk of recurrence is histology-specific. Following multimodality treatment of the skull base, patients may experience endocrine, visual, auditory, sinonasal, olfactory, and neurocognitive deficits, as well as psychosocial impairments that impact multiple physical and neuropsychological domains, resulting in diminished quality of life. Sinonasal cancer patients would benefit from tailored, histology-specific survivorship programs to address the recurrence, second primary, and functional impairments resulting from disease and treatment toxicity.
Collapse
Affiliation(s)
- Albert Y Han
- Department of Head and Neck Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Marc-Elie Nader
- Department of Head and Neck Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Keng Lam
- Department of Neuro-Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Shirley Y Su
- Department of Head and Neck Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
10
|
Waissbluth S, Martínez AD, Figueroa-Cares C, Sánchez HA, Maass JC. MATE1 expression in the cochlea and its potential involvement in cisplatin cellular uptake and ototoxicity. Acta Otolaryngol 2023; 143:242-249. [PMID: 36943799 DOI: 10.1080/00016489.2023.2184864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
BACKGROUND Cisplatin appears to enter the cochlear cells through the organic cation transporter 2 (OCT2). There is recent evidence that multidrug and toxin extrusion protein 1 (MATE1) is involved in cisplatin-induced nephrotoxicity. Its presence and role in the ear are unknown. AIMS/OBJECTIVES Evaluate the presence and localization of MATE1, and determine the localization of OCT2, in the cochlea. Evaluate cisplatin uptake with regard to MATE1 and OCT2 expression. MATERIAL AND METHODS Murine cochlear explants and paraffin-embedded cochleae were evaluated with immunohistochemistry for OCT2 and MATE1. Explant cultures were also treated with Texas Red cisplatin to determine their cellular uptake. RESULTS MATE1 is present in the cochlea. Most intense labeling of MATE1 and OCT2 was seen in the outer hair cells (OHCs) and pillar cells, respectively. Both transporters were observed in the spiral ganglion neurons and stria vascularis. Expression levels of OCT2 and MATE1 decreased following cisplatin exposure. Texas Red cisplatin staining was strong in OHCs and pillar cells. CONCLUSIONS AND SIGNIFICANCE To the best of our knowledge, this is the first study demonstrating the presence and localization of MATE1 in the cochlea. OCT2 labeling was seen in pillar cells. Consistently, OHCs and pillar cells uptake Texas Red cisplatin.
Collapse
Affiliation(s)
- Sofia Waissbluth
- Department of Otolaryngology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Cindel Figueroa-Cares
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Helmuth A Sánchez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Juan C Maass
- Department of Otolaryngology, Hospital Clínico Universidad de Chile and Interdisciplinary Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Surgery, Clínica Alemana de Santiago, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
DeBacker JR, McMillan GP, Martchenke N, Lacey CM, Stuehm HR, Hungerford ME, Konrad-Martin D. Ototoxicity prognostic models in adult and pediatric cancer patients: a rapid review. J Cancer Surviv 2023; 17:82-100. [PMID: 36729346 PMCID: PMC11727884 DOI: 10.1007/s11764-022-01315-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/07/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE A cornerstone of treatment for many cancers is the administration of platinum-based chemotherapies and/or ionizing radiation, which can be ototoxic. An accurate ototoxicity risk assessment would be useful for counseling, treatment planning, and survivorship follow-up in patients with cancer. METHODS This systematic review evaluated the literature on predictive models for estimating a patient's risk for chemotherapy-related auditory injury to accelerate development of computational approaches for the clinical management of ototoxicity in cancer patients. Of the 1195 articles identified in a PubMed search from 2010 forward, 15 studies met inclusion for the review. CONCLUSIONS All but 1 study used an abstraction of the audiogram as a modeled outcome; however, specific outcome measures varied. Consistently used predictors were age, baseline hearing, cumulative cisplatin dose, and radiation dose to the cochlea. Just 5 studies were judged to have an overall low risk of bias. Future studies should attempt to minimize bias by following statistical best practices including not selecting multivariate predictors based on univariate analysis, validation in independent cohorts, and clearly reporting the management of missing and censored data. Future modeling efforts should adopt a transdisciplinary approach to define a unified set of clinical, treatment, and/or genetic risk factors. Creating a flexible model that uses a common set of predictors to forecast the full post-treatment audiogram may accelerate work in this area. Such a model could be adapted for use in counseling, treatment planning, and follow-up by audiologists and oncologists and could be incorporated into ototoxicity genetic association studies as well as clinical trials investigating otoprotective agents. IMPLICATIONS FOR CANCER SURVIVORS Improvements in the ability to model post-treatment hearing loss can help to improve patient quality of life following cancer care. The improvements advocated for in this review should allow for the acceleration of advancements in modeling the auditory impact of these treatments to support treatment planning and patient counseling during and after care.
Collapse
Affiliation(s)
- J R DeBacker
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA.
- Oregon Health and Science University, Portland, OR, USA.
| | - G P McMillan
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA
- Oregon Health and Science University, Portland, OR, USA
| | - N Martchenke
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA
- Oregon Health and Science University, Portland, OR, USA
| | - C M Lacey
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA
- University of Pittsburgh, Pittsburgh, PA, USA
| | - H R Stuehm
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA
- Oregon Health and Science University, Portland, OR, USA
| | - M E Hungerford
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA
- Oregon Health and Science University, Portland, OR, USA
| | - D Konrad-Martin
- VA RR&D National Center for Rehabilitative Auditory Research, VA Portland Health Care System, 3710 SW US Veterans Hospital Road (NCRAR - P5), Portland, OR, 97239, USA
- Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
14
|
Lee DS, Mahal RS, Tharakan T, Cathryn Collopy, Kallogjeri D, Thorstad WL, Adkins DR, Oppelt P, Ley J, Wick CC, Zevallos J. Hearing Outcomes in a Deintensification Trial of Adjuvant Therapy for HPV-Related Oropharyngeal Squamous Cell Carcinoma. Otolaryngol Head Neck Surg 2023; 168:1089-1096. [PMID: 36939390 DOI: 10.1002/ohn.182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/27/2022] [Accepted: 10/08/2022] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To explore whether deintensification of adjuvant therapy reduces ototoxicity among patients with human papillomavirus (HPV)-related oropharyngeal squamous cell carcinoma (OPSCC). STUDY DESIGN Retrospective cohort study. SETTING Single academic center. METHODS The ototoxicity rate among adult patients with HPV-related OPSCC enrolled in the Minimalist Trial (MINT), a prospective phase 2 trial of surgery followed by risk-adjusted deintensified adjuvant therapy (42 Gy radiation given alone or with a single 100 mg/m2 dose of cisplatin), was compared to that among a historical cohort treated with standard adjuvant therapy (60-66 Gy radiation with up to three 100 mg/m2 doses of cisplatin). Ototoxicity was defined as Common Terminology Criteria for Adverse Events v5.0 ≥ Grade 2. Mixed model analysis was performed to investigate the association between deintensified adjuvant therapy and treatment-related hearing loss. RESULTS A total of 29 patients (58 ears) were analyzed in the MINT cohort, and 27 patients (54 ears) in the historical cohort. The ototoxicity rate was 5% (n = 3/58 ears) in the MINT cohort and 46% (n = 25/54 ears) in the historical cohort (difference, 41%; 95% confidence interval [CI] = 27%-56%). Patients in the MINT cohort demonstrated a 95% decrease in risk of ototoxicity compared to those in the historical cohort (adjusted odds ratio: 0.05, 95% CI = 0.01-0.31). Differences in estimated marginal mean threshold shifts were statistically and clinically significant at frequencies ≥ 3 kHz. CONCLUSION The deintensified adjuvant therapy given in MINT led to less ototoxicity than standard adjuvant therapy among patients with HPV-related OPSCC.
Collapse
Affiliation(s)
- David S Lee
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Rajwant S Mahal
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Theresa Tharakan
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Cathryn Collopy
- Department of Otolaryngology-Head and Neck Surgery, Division of Adult Audiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Dorina Kallogjeri
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Wade L Thorstad
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Douglas R Adkins
- Department of Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Peter Oppelt
- Department of Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jessica Ley
- Department of Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Cameron C Wick
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jose Zevallos
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
15
|
Okada H, Kitagawa K. Risk factors associated with cisplatin-induced ototoxicity in Japanese patients with solid tumors. Cancer Med 2022; 12:7904-7910. [PMID: 36567514 PMCID: PMC10134260 DOI: 10.1002/cam4.5565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cisplatin, a first-generation platinum agent, is used for managing various cancers and is associated with dose-dependent side effects of hearing impairment and tinnitus. However, the safety of high-dose cisplatin in hearing impairment, has not been fully investigated in Japan. METHODS We performed pure-tone threshold audiometry before and every 3-4 weeks after chemotherapy for patients receiving cisplatin-containing chemotherapy between April 2015 and October 2017 at Kobe Minimally Invasive Cancer Center. Hearing impairment was evaluated prospectively using the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. RESULTS We enrolled 100 patients and analyzed 96 patients for whom post-chemotherapy audiometry could be performed. The median patient age was 65 years, and most patients were male (75). The cancer types were as follows: esophageal, 36; head and neck, 35; lung, 23; and gastric, 2. Cisplatin monotherapy and combination therapy were administered to 33 and 63 patients, respectively. A single cisplatin dose was 60-100 mg/m2 ; the median number of doses and total dose were 3 and 240 mg/m2 , respectively. Additionally, 78 and 18 patients were treated with concurrent chemoradiotherapy and chemotherapy alone, respectively. Twenty-seven patients had grade 2 or higher hearing impairment. Furthermore, the prevalence was significantly higher in patients receiving a total dose of ≥300 mg/m2 . Twenty and 32 patients were aware of deafness and tinnitus, respectively. CONCLUSION No patient discontinued treatment owing to hearing impairment. The total cisplatin dose was considered related to post-treatment hearing impairment frequency in Japanese patients. However, routine audiometric monitoring is recommended during high-dose cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Hideaki Okada
- Division of Medical Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Japan
| | - Koichi Kitagawa
- Division of Medical Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Japan
| |
Collapse
|
16
|
Zeng PYF, Cecchini MJ, Barrett JW, Shammas-Toma M, De Cecco L, Serafini MS, Cavalieri S, Licitra L, Hoebers F, Brakenhoff RH, Leemans CR, Scheckenbach K, Poli T, Wang X, Liu X, Laxague F, Prisman E, Poh C, Bose P, Dort JC, Shaikh MH, Ryan SEB, Dawson A, Khan MI, Howlett CJ, Stecho W, Plantinga P, Daniela da Silva S, Hier M, Khan H, MacNeil D, Mendez A, Yoo J, Fung K, Lang P, Winquist E, Palma DA, Ziai H, Amelio AL, Li SSC, Boutros PC, Mymryk JS, Nichols AC. Immune-based classification of HPV-associated oropharyngeal cancer with implications for biomarker-driven treatment de-intensification. EBioMedicine 2022; 86:104373. [PMID: 36442320 PMCID: PMC9706534 DOI: 10.1016/j.ebiom.2022.104373] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND There is significant interest in treatment de-escalation for human papillomavirus-associated (HPV+) oropharyngeal squamous cell carcinoma (OPSCC) patients given the generally favourable prognosis. However, 15-30% of patients recur after primary treatment, reflecting a need for improved risk-stratification tools. We sought to develop a molecular test to risk stratify HPV+ OPSCC patients. METHODS We created an immune score (UWO3) associated with survival outcomes in six independent cohorts comprising 906 patients, including blinded retrospective and prospective external validations. Two aggressive radiation de-escalation cohorts were used to assess the ability of UWO3 to identify patients who recur. Multivariate Cox models were used to assess the associations between the UWO3 immune class and outcomes. FINDINGS A three-gene immune score classified patients into three immune classes (immune rich, mixed, or immune desert) and was strongly associated with disease-free survival in six datasets, including large retrospective and prospective datasets. Pooled analysis demonstrated that the immune rich group had superior disease-free survival compared to the immune desert (HR = 9.0, 95% CI: 3.2-25.5, P = 3.6 × 10-5) and mixed (HR = 6.4, 95% CI: 2.2-18.7, P = 0.006) groups after adjusting for age, sex, smoking status, and AJCC8 clinical stage. Finally, UWO3 was able to identify patients from two small treatment de-escalation cohorts who remain disease-free after aggressive de-escalation to 30 Gy radiation. INTERPRETATION With additional prospective validation, the UWO3 score could enable biomarker-driven clinical decision-making for patients with HPV+ OPSCC based on robust outcome prediction across six independent cohorts. Prospective de-escalation and intensification clinical trials are currently being planned. FUNDING CIHR, European Union, and the NIH.
Collapse
Affiliation(s)
- Peter Y F Zeng
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - John W Barrett
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Matthew Shammas-Toma
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Loris De Cecco
- Integrated Biology Platform, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy
| | - Mara S Serafini
- Integrated Biology Platform, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), Research Institute GROW, Maastricht University, Maastricht, the Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, the Netherlands
| | - C René Leemans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, the Netherlands
| | - Kathrin Scheckenbach
- Department of Otolaryngology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tito Poli
- Unit of Maxillofacial Surgery, Department of Medicine and Surgery, University of Parma-University Hospital of Parma, Parma, Italy
| | - Xiaowei Wang
- Department of Pharmacology and Regenerative Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Xinyi Liu
- Department of Pharmacology and Regenerative Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Francisco Laxague
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Eitan Prisman
- Division of Otolaryngology- Head and Neck Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Catherine Poh
- Division of Otolaryngology- Head and Neck Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, British Columbia, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pinaki Bose
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Joseph C Dort
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Mushfiq H Shaikh
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Sarah E B Ryan
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Alice Dawson
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Mohammed I Khan
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Christopher J Howlett
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - William Stecho
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Paul Plantinga
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | | | - Michael Hier
- Department of Otolaryngology Head and Neck Surgery, McGill University, Montreal, Quebec, Canada
| | - Halema Khan
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Danielle MacNeil
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Adrian Mendez
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - John Yoo
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Kevin Fung
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Pencilla Lang
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Eric Winquist
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - David A Palma
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Hedyeh Ziai
- Department of Otolaryngology - Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Antonio L Amelio
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn S-C Li
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA; Department of Urology, University of California, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA; Institute for Precision Health, University of California, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, CA, USA
| | - Joe S Mymryk
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada; Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
17
|
Noman A, Mukherjee S, Le TN. Manipulating the Blood Labyrinth Barrier with Mannitol to Prevent Cisplatin-Induced Hearing Loss. Hear Res 2022; 426:108646. [DOI: 10.1016/j.heares.2022.108646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/10/2022] [Accepted: 11/04/2022] [Indexed: 11/08/2022]
|
18
|
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
|
19
|
Marcu LG. Gender and Sex-Related Differences in Normal Tissue Effects Induced by Platinum Compounds. Pharmaceuticals (Basel) 2022; 15:255. [PMID: 35215367 PMCID: PMC8876358 DOI: 10.3390/ph15020255] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/18/2022] Open
Abstract
Gender medicine in the field of oncology is an under-researched area, despite the existing evidence towards gender-dependent response to therapy and treatment-induced adverse effects. Oncological treatment aims to fulfil its main goal of achieving high tumour control by also protecting normal tissue from acute or chronic damage. Chemotherapy is an important component of cancer treatment, with a large number of drugs being currently in clinical use. Cisplatin is one of the most commonly employed chemotherapeutic agents, used either as a sole drug or in combination with other agents. Cisplatin-induced toxicities are well documented, and they include nephrotoxicity, neurotoxicity, gastrointestinal toxicity, ototoxicity, just to name the most frequent ones. Some of these toxicities have short-term sequelae, while others are irreversible. Furthermore, research showed that there is a strong gender-dependent aspect of side effects caused by the administration of cisplatin. While evidence towards sex differences in animal models is substantial, clinical studies considering sex/gender as a variable factor are limited. This work summarises the current knowledge on sex/gender-related side effects induced by platinum compounds and highlights the gaps in research that require more attention to open new therapeutic possibilities and preventative measures to alleviate normal tissue toxicity and increase patients' quality of life in both males and females.
Collapse
Affiliation(s)
- Loredana G. Marcu
- Faculty of Informatics & Science, Department of Physics, University of Oradea, 410087 Oradea, Romania;
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
20
|
Abstract
Pharmacogenetic testing in patients with cancer requiring cytotoxic chemotherapy offers the potential to predict, prevent, and mitigate chemotherapy-related toxicities. While multiple drug-gene pairs have been identified and studied, few drug-gene pairs are currently used routinely in the clinical status. Here we review what is known, theorized, and unknown regarding the use of pharmacogenetic testing in cancer.
Collapse
Affiliation(s)
- Zahra Talebi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Susan I Colace
- Division of Hematology, Oncology, and Blood & Marrow Transplant, Nationwide Children's Hospital, Columbus, OH, USA.
- The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
21
|
Konrad-Martin D, O'Connell Bennett K, Garinis A, McMillan GP. A Randomized Controlled Trial Using Automated Technology for Improving Ototoxicity Monitoring in VA Oncology Patients. Am J Audiol 2021; 30:870-886. [PMID: 34582263 DOI: 10.1044/2021_aja-21-00032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Purpose Determine the efficacy of ototoxicity monitoring (OM) administered as automated protocols with the Oto-ID mobile audiometer (automated ototoxicity monitoring [A-OM]), compared with usual care (UC) OM in cancer patients receiving cisplatin. Method Participants were patients (n = 46, mean age 64.7 years; range: 30-78 years) receiving cisplatin-based chemotherapy at the Department of Veterans Affairs Portland Health Care System. A randomized controlled trial contrasted A-OM and UC at up to three program evaluations (PEs) conducted by the study audiologist who was blinded to arm through PE1. PE1 occurred before randomization or oncology treatment; PE2 and PE3 occurred during and/or after treatment at 35 and 365 days postrandomization. The A-OM group (n = 24) used Oto-ID to screen their hearing before each cisplatin dose. Oto-ID results were sent to the study audiologist for interpretation, follow-up, and care coordination. The UC group (n = 22) received a consult for OM services through the audiology clinic. Outcomes included hearing shift near each patient's high-frequency hearing limit, revised hearing-handicap inventory score, and survival time from the start of treatment. Adherence to OM protocols, patients' use of aural rehabilitation services, and oncologists' treatment decisions were also examined. Results Ototoxicity was identified at a high overall rate (46% and 76% at 35 and 365 days, respectively, postrandomization). Adherence to monitoring prior to each cisplatin dose was 83.3% for those randomized to A-OM compared with 4.5% for UC. Randomization to A-OM was not associated with reduced ototoxic hearing shifts or self-reported hearing handicap relative to UC; neither did it compromise participants' survival. Half of participants in each arm accessed aural rehabilitation services. One in each arm had a documented ototoxicity-related cisplatin dose reduction. Conclusions Auditory impairment was an actionable concern for the participants and their oncology providers. A dedicated surveillance program using the Oto-ID's automated protocols improved adherence to OM recommendations over a traditional UC service delivery model. Supplemental Material https://doi.org/10.23641/asha.16649602.
Collapse
Affiliation(s)
- Dawn Konrad-Martin
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, OR
- Department of Otolaryngology—Head & Neck Surgery, Oregon Health & Science University, Portland
| | - Keri O'Connell Bennett
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, OR
| | - Angela Garinis
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, OR
- Department of Otolaryngology—Head & Neck Surgery, Oregon Health & Science University, Portland
- Oregon Hearing Research Center, Oregon Health & Science University, Portland
| | - Garnett P. McMillan
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, OR
- Department of Otolaryngology—Head & Neck Surgery, Oregon Health & Science University, Portland
| |
Collapse
|
22
|
Santucci NM, Garber B, Ivory R, Kuhn MA, Stephen M, Aizenberg D. Insight into the current practice of ototoxicity monitoring during cisplatin therapy. J Otolaryngol Head Neck Surg 2021; 50:19. [PMID: 33766142 PMCID: PMC7995701 DOI: 10.1186/s40463-021-00506-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/22/2021] [Indexed: 11/20/2022] Open
Abstract
Background The aim of this study is to evaluate the current state of ototoxicity monitoring for patients receiving cisplatin chemotherapy in an academic medical center with particular attention to how closely monitoring adheres to national ototoxicity guidelines. Methods Case series including retrospective medical records review of patients (age > 18) treated with cisplatin at University of California Davis Medical Center between January 2014 and August 2017. Patient and ototoxicity related variables were analyzed. Patients that underwent a transfer of care during treatment and with less than 3 months of follow-up were excluded. Results Three hundred seventy-nine patients met study criteria, of which 104 (27.4%) had a prior history of hearing loss. Prior to treatment, 196 (51.7%) patients were counseled regarding the ototoxic nature of cisplatin and 92 (24.3%) patients had a pretreatment audiogram. During treatment, 91 (24%) patients had documented otologic complaints. Only 17 patients (4.5%) patients had an audiogram ordered during their cisplatin treatment period. 130 (34.3%) patients had otologic complaints following cisplatin treatment. Audiograms were ordered for 20 (7.8%), 13 (5.1%), and 16 (6.2%) patients at 1-month, 3-month, and 6-month follow-ups, respectively. No patients in the study cohort received baseline, treatment, and post-treatment audiograms as recommended by national ototoxicity monitoring protocols. Patients with Head and Neck Cancer (HNC) represented the largest subgroup that received cisplatin (n = 122, 32.2%) and demonstrated higher rates of ototoxicity counseling (n = 103, 84.4%) and pretreatment audiograms (n = 70, 57.4%) compared to the non HNC group (n = 36, 36.2%, P < 0.0001 and n = 22, 8.5%, P < 0.0001). Conclusions There is poor adherence to national ototoxicity monitoring guidelines at a large academic medical center. This is a missed opportunity for intervention and aural rehabilitation. Improved education and collaboration between otolaryngology, audiology, and medical oncology is needed to develop and promote an effective ototoxicity-monitoring program. Graphical abstract ![]()
Collapse
Affiliation(s)
- N M Santucci
- Oregon Health and Science University, School of Medicine, Portland, OR, USA
| | - B Garber
- University of California Davis Department of Otolaryngology-Head and Neck Surgery, 2521 Stockton Blvd., Sacramento, CA, 95817, USA
| | - R Ivory
- University of California Davis Medical Center, Sacramento, CA, USA
| | - M A Kuhn
- University of California Davis Department of Otolaryngology-Head and Neck Surgery, 2521 Stockton Blvd., Sacramento, CA, 95817, USA
| | - M Stephen
- University of California Davis Department of Internal Medicine - Hematology/Oncology, Sacramento, CA, USA
| | - D Aizenberg
- University of California Davis Department of Otolaryngology-Head and Neck Surgery, 2521 Stockton Blvd., Sacramento, CA, 95817, USA.
| |
Collapse
|
23
|
Shim HJ, Kim HJ, Hwang JE, Bae WK, Chung IJ, Lee DH, Mi YT, Lee JK, Lim SC, Chung JW, Cho SH. Long term complications and prognostic factors in locally advanced nasopharyngeal carcinoma treated with docetaxel, cisplatin, 5-fluorouracil induction chemotherapy followed by concurrent chemoradiotherapy: A retrospective cohort study. Medicine (Baltimore) 2020; 99:e23173. [PMID: 33285692 PMCID: PMC7717786 DOI: 10.1097/md.0000000000023173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
This study was conducted to evaluate the long term complications and their risk factors including of survival outcomes in patients with locally advanced nasopharyngeal cancer (NPC) treated with docetaxel, cisplatin and 5-fluorouracil (TPF) induction chemotherapy followed by concurrent chemoradiotherapy (CCRT).Among the patients who were diagnosed as NPC, we consecutively evaluated the late complications in 104 patients who completed 3 cycles of TPF induction chemotherapy followed by CCRT and received regular follow-up by otolaryngologist and oncologist. The prognostic factors for overall survival, relapse free survival and each complication were analyzed based on clinical characteristics.Over a median follow-up of 54 months (range, 7.9-152.9 months), 5-year overall survival rate was 87% for stage II, 89% for stage III, 87% for stage IV patients. The significant prognostic factor for survival is complete response rate after CCRT in multivariate analysis. The most frequent toxicity was ear complication (29.8%) including of hearing loss requiring hearing aid (6.7%) and bone necrosis (3.8%). Decreased renal function over grade 2 was occurred in only 4 patients (3.8%) regardless of the cumulative dose of cisplatin. The long term complications did not affect the survival outcome. Patients who received radiation therapy more than 5400 cGy had better survival outcome than those who did not. However, ear complication was significantly related to radiation dose (≥ 6,600 cGy) and type of radiation therapy (conventional). Age over 65 years was a significant risk factor for both ear and renal toxicity. In conclusion, close follow-up to monitor long-term complications should be performed in patients treated with TPF induction chemotherapy followed by CCRT treatment, especially in elderly patients. Reestablishing the optimal chemotherapeutic agent during CCRT and adjustment of radiation dose after induction chemotherapy could be helpful to reduce the toxicity associated with the subsequent treatment strategy for locally advance NPC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jae Wook Chung
- Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | | |
Collapse
|
24
|
Zazuli Z, Duin NJCB, Jansen K, Vijverberg SJH, Maitland-van der Zee AH, Masereeuw R. The Impact of Genetic Polymorphisms in Organic Cation Transporters on Renal Drug Disposition. Int J Mol Sci 2020; 21:ijms21186627. [PMID: 32927790 PMCID: PMC7554776 DOI: 10.3390/ijms21186627] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
A considerable number of drugs and/or their metabolites are excreted by the kidneys through glomerular filtration and active renal tubule secretion via transporter proteins. Uptake transporters in the proximal tubule are part of the solute carrier (SLC) superfamily, and include the organic cation transporters (OCTs). Several studies have shown that specific genetic polymorphisms in OCTs alter drug disposition and may lead to nephrotoxicity. Multiple single nucleotide polymorphisms (SNPs) have been reported for the OCT genes (SLC22A1, SLC22A2 and SLC22A3), which can influence the proteins’ structure and expression levels and affect their transport function. A gain-in-function mutation may lead to accumulation of drugs in renal proximal tubule cells, eventually leading to nephrotoxicity. This review illustrates the impact of genetic polymorphisms in OCTs on renal drug disposition and kidney injury, the clinical significances and how to personalize therapies to minimize the risk of drug toxicity.
Collapse
Affiliation(s)
- Zulfan Zazuli
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.J.H.V.); (A.H.M.-v.d.Z.)
- Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Bandung Institute of Technology, Jawa Barat 40132, Indonesia
- Correspondence: (Z.Z.); (R.M.)
| | - Naut J. C. B. Duin
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.J.C.B.D.); (K.J.)
| | - Katja Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.J.C.B.D.); (K.J.)
| | - Susanne J. H. Vijverberg
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.J.H.V.); (A.H.M.-v.d.Z.)
| | - Anke H. Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.J.H.V.); (A.H.M.-v.d.Z.)
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.J.C.B.D.); (K.J.)
- Correspondence: (Z.Z.); (R.M.)
| |
Collapse
|
25
|
Langer T, Clemens E, Broer L, Maier L, Uitterlinden AG, de Vries ACH, van Grotel M, Pluijm SFM, Binder H, Mayer B, von dem Knesebeck A, Byrne J, van Dulmen-den Broeder E, Crocco M, Grabow D, Kaatsch P, Kaiser M, Spix C, Kenborg L, Winther JF, Rechnitzer C, Hasle H, Kepak T, van der Kooi ALF, Kremer LC, Kruseova J, Bielack S, Sorg B, Hecker-Nolting S, Kuehni CE, Ansari M, Kompis M, van der Pal H, Parfitt R, Deuster D, Matulat P, Tillmanns A, Tissing WJE, Beck JD, Elsner S, Am Zehnhoff-Dinnesen A, van den Heuvel-Eibrink MM, Zolk O. Usefulness of current candidate genetic markers to identify childhood cancer patients at risk for platinum-induced ototoxicity: Results of the European PanCareLIFE cohort study. Eur J Cancer 2020; 138:212-224. [PMID: 32905960 DOI: 10.1016/j.ejca.2020.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND Irreversible sensorineural hearing loss is a common side effect of platinum treatment with the potential to significantly impair the neurocognitive, social and educational development of childhood cancer survivors. Genetic association studies suggest a genetic predisposition for cisplatin-induced ototoxicity. Among other candidate genes, thiopurine methyltransferase (TPMT) is considered a critical gene for susceptibility to cisplatin-induced hearing loss in a pharmacogenetic guideline. The aim of this cross-sectional cohort study was to confirm the genetic associations in a large pan-European population and to evaluate the diagnostic accuracy of the genetic markers. METHODS Eligibility criteria required patients to be aged less than 19 years at the start of chemotherapy, which had to include cisplatin and/or carboplatin. Patients were assigned to three phenotype categories: no, minor and clinically relevant hearing loss. Fourteen variants in eleven candidate genes (ABCC3, OTOS, TPMT, SLC22A2, NFE2L2, SLC16A5, LRP2, GSTP1, SOD2, WFS1 and ACYP2) were investigated. Multinomial logistic regression was performed to model the relationship between genetic predictors and platinum ototoxicity, adjusting for clinical risk factors. Additionally, measures of the diagnostic accuracy of the genetic markers were determined. RESULTS 900 patients were included in this study. In the multinomial logistic regression, significant unique contributions were found from SLC22A2 rs316019, the age at the start of platinum treatment, cranial radiation and the interaction term [platinum compound]∗[cumulative dose of cisplatin]. The predictive performance of the genetic markers was poor compared with the clinical risk factors. CONCLUSIONS PanCareLIFE is the largest study of cisplatin-induced ototoxicity to date and confirmed a role for the polyspecific organic cation transporter SLC22A2. However, the predictive value of the current genetic candidate markers for clinical use is negligible, which puts the value of clinical factors for risk assessment of cisplatin-induced ototoxicity back into the foreground.
Collapse
Affiliation(s)
- Thorsten Langer
- Department of Pediatric Oncology and Hematology, University Hospital for Children and Adolescents, Lübeck, Germany
| | - Eva Clemens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Linda Broer
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Lara Maier
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University Medical Center, Ulm, Germany
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Andrica 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
| | | | - Saskia F M Pluijm
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Harald 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
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Annika von dem Knesebeck
- Department of Pediatric Oncology and Hematology, University Hospital for Children and Adolescents, Lübeck, Germany
| | | | - Eline van Dulmen-den Broeder
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Hematology and Oncology, VU Medical Center, Amsterdam, the Netherlands
| | - Marco Crocco
- Department of Neurooncology, Istituto Giannina Gaslini, Genova, Italy
| | - Desiree Grabow
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Peter Kaatsch
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie Kaiser
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Spix
- German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Line Kenborg
- Danish Cancer Society Research Center, Childhood Cancer Research Group, Copenhagen, Denmark
| | - Jeanette F Winther
- Danish Cancer Society Research Center, Childhood Cancer Research Group, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Catherine Rechnitzer
- Copenhagen University Hospital Rigshospitalet, Department of Pediatrics and Adolescent Medicine, Copenhagen, Denmark
| | - Henrik Hasle
- Aarhus University Hospital, Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | - Tomas Kepak
- University Hospital Brno, Brno, Czech Republic; International Clinical Research Center (FNUSA-ICRC), Brno, Czech Republic
| | - Anne-Lotte F van der Kooi
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Obstetrics and Gynecology, Erasmus MC - Sophia Children's Hospital, the Netherlands
| | - Leontien C Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Oncology, Academic Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Jarmila Kruseova
- Department of Children Hemato-Oncology, Motol University Hospital Prague, Prague, Czech Republic
| | - Stefan Bielack
- Department of Pediatric Oncology, Hematology, Immunology, Stuttgart Cancer Center, Olgahospital, Stuttgart, Germany
| | - Benjamin Sorg
- Department of Pediatric Oncology, Hematology, Immunology, Stuttgart Cancer Center, Olgahospital, Stuttgart, Germany
| | - Stefanie Hecker-Nolting
- Department of Pediatric Oncology, Hematology, Immunology, Stuttgart Cancer Center, Olgahospital, Stuttgart, Germany
| | - Claudia E Kuehni
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland; Paediatric Oncology, Dept. of Paediatrics, Inselspital, University of Bern, Switzerland
| | - Marc Ansari
- Department of Pediatrics, Oncology and Hematology Unit, University Hospital of Geneva, Cansearch Research Laboratory, Geneva University, Switzerland
| | - Martin Kompis
- Department of Otolaryngology, Head and Neck Surgery, Inselspital, University of Berne, Switzerland
| | - Heleen van der Pal
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Oncology, Academic Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Ross Parfitt
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - Dirk Deuster
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - Peter Matulat
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - Amelie Tillmanns
- Department of Phoniatrics and Pedaudiology, University Hospital Münster, Westphalian Wilhelm University, Münster, Germany
| | - Wim 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
| | - Jörn D Beck
- Hospital for Children and Adolescents, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Susanne Elsner
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | | | - Marry 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
| | - Oliver Zolk
- Institute of Clinical Pharmacology, Immanuel Klinik Rüdersdorf, Brandenburg Medical School Theodor Fontane, Germany; Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University Medical Center, Ulm, Germany.
| | | |
Collapse
|
26
|
Chen Y, Zhou H, Yang S, Su D. Increased ABCC2 expression predicts cisplatin resistance in non-small cell lung cancer. Cell Biochem Funct 2020; 39:277-286. [PMID: 32815556 PMCID: PMC7983913 DOI: 10.1002/cbf.3577] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/26/2020] [Accepted: 07/05/2020] [Indexed: 12/23/2022]
Abstract
Long-term use of platinum-based drugs can cause non-small cell lung cancer (NSCLC) to develop extremely strong drug resistance. Increasing the drug dosage does not have better treatment effects and could lead to serious complications. High levels of drug resistance are considered to be characteristic of human tumours and are usually mediated by genes related to multidrug resistance. Multidrug resistance-associated protein 2 (ABCC2), an ATP-binding cassette multidrug resistance transporter, was found to be overexpressed in various human cancers. In this study, we found that ABCC2 was also upregulated in cisplatin (DDP)-resistant A549 cells (A549/DDP). Functional studies demonstrated that ABCC2 knockdown reversed DDP resistance and promoted G1 phase arrest in A549/DDP cells, and PARP and caspase-3 were activated in A549/DDP cells following ABCC2 knockdown. In vivo, ABCC2 knockdown enhanced the cytotoxicity of DDP to subcutaneous A549 tumours. Together, these results suggest that ABCC2 may be a potential therapeutic strategy for overcoming DDP resistance in NSCLC patients. SIGNIFICANCE OF THE STUDY: In this study, we investigated the role of ABCC2 in cisplatin resistance of NSCLC cells. Our data show that ABCC2 expression was associated with resistance to cisplatin and that knockdown ABCC2 could reverse cisplatin resistance in NSCLC cells. Taken together, our study suggests that reducing the expression of ABCC2 could become an important strategy for enhancing the sensitivity of NSCLC cells to cisplatin.
Collapse
Affiliation(s)
- Yun Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Hongying Zhou
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Sifu Yang
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Dan Su
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
27
|
Huang KM, Uddin ME, DiGiacomo D, Lustberg MB, Hu S, Sparreboom A. Role of SLC transporters in toxicity induced by anticancer drugs. Expert Opin Drug Metab Toxicol 2020; 16:493-506. [PMID: 32276560 DOI: 10.1080/17425255.2020.1755253] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION . Membrane transporters are integral to the maintenance of cellular integrity of all tissue and cell types. While transporters play an established role in the systemic pharmacokinetics of therapeutic drugs, tissue specific expression of uptake transporters can serve as an initiating mechanism that governs the accumulation and impact of cytotoxic drugs. AREAS COVERED . This review provides an overview of organic cation transporters as determinants of chemotherapy-induced toxicities. We also provide insights into the recently updated FDA guidelines for in vitro drug interaction studies, with a particular focus on the class of tyrosine kinase inhibitors as perpetrators of transporter-mediated drug interactions. EXPERT OPINION . Studies performed over the last few decades have highlighted the important role of basolateral uptake and apical efflux transporters in the pathophysiology of drug-induced organ damage. Increased understanding of the mechanisms that govern the accumulation of cytotoxic drugs has provided insights into the development of novel strategies to prevent debilitating toxicities. Furthermore, we argue that current regulatory guidelines provide inadequate recommendations for in vitro studies to identify substrates or inhibitors of drug transporters. Therefore, the translational and predictive power of FDA-approved drugs as modulators of transport function remains ambiguous and warrants further revision of the current guidelines.
Collapse
Affiliation(s)
- Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Muhammad Erfan Uddin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Duncan DiGiacomo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Maryam B Lustberg
- Department of Medical Oncology, College of Medicine, the Ohio State University and Comprehensive Cancer Center , Columbus, OH, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| |
Collapse
|
28
|
DeBacker JR, Harrison RT, Bielefeld EC. Cisplatin-induced threshold shift in the CBA/CaJ, C57BL/6J, BALB/cJ mouse models of hearing loss. Hear Res 2020; 387:107878. [DOI: 10.1016/j.heares.2019.107878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 10/25/2022]
|
29
|
Palma DA, Mitchell S, Nichols A. Sparing of swallowing-related organs in radiotherapy for oropharyngeal squamous cell carcinoma-Authors' reply. Lancet Oncol 2019; 20:e612. [PMID: 31674316 DOI: 10.1016/s1470-2045(19)30653-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 11/19/2022]
Affiliation(s)
- David A Palma
- London Health Sciences Centre, London, ON N6A 5W9, Canada.
| | | | | |
Collapse
|
30
|
Olivera G, Sendra L, Herrero MJ, Puig C, Aliño SF. Colorectal cancer: pharmacogenetics support for the correct drug prescription. Pharmacogenomics 2019; 20:741-763. [PMID: 31368847 DOI: 10.2217/pgs-2019-0041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pharmacogenetics (PGx) in clinical practice is a tool that the clinician can use to guide, in a personalized way, the most suitable treatment that will be administered to the patient. The objective of this review is to summarize in a practical and accessible rational way, the advances that currently exist for the application of PGx in colorectal cancer chemotherapy management through the study of the patients' germline polymorphisms. To define the polymorphisms that can be applied, we rely on three fundamental cornerstones: the recommendations of drug regulatory agencies; the implementation guidelines prepared by expert consortia in PGx and information from clinical annotations (the drug/polymorphism relation) according to the scientific level of evidence assigned by PharmGKB experts.
Collapse
Affiliation(s)
- Gladys Olivera
- Pharmacogenetics Platform, Instituto de Investigación Sanitaria la Fe, Valencia 46026, Spain.,Department of Pharmacology, University of Valencia, Valencia 46010, Spain
| | - Luis Sendra
- Pharmacogenetics Platform, Instituto de Investigación Sanitaria la Fe, Valencia 46026, Spain.,Department of Pharmacology, University of Valencia, Valencia 46010, Spain
| | - María José Herrero
- Pharmacogenetics Platform, Instituto de Investigación Sanitaria la Fe, Valencia 46026, Spain.,Department of Pharmacology, University of Valencia, Valencia 46010, Spain
| | - Carlos Puig
- Department of Pharmacology, University of Valencia, Valencia 46010, Spain
| | - Salvador F Aliño
- Pharmacogenetics Platform, Instituto de Investigación Sanitaria la Fe, Valencia 46026, Spain.,Department of Pharmacology, University of Valencia, Valencia 46010, Spain.,Clinical Pharmacology Unit, Hospital Universitario y Politécnico la Fe, Valencia 46026, Spain
| |
Collapse
|
31
|
Drögemöller BI, Wright GE, Lo C, Le T, Brooks B, Bhavsar AP, Rassekh SR, Ross CJ, Carleton BC. Pharmacogenomics of Cisplatin‐Induced Ototoxicity: Successes, Shortcomings, and Future Avenues of Research. Clin Pharmacol Ther 2019; 106:350-359. [DOI: 10.1002/cpt.1483] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Britt I. Drögemöller
- Faculty of Pharmaceutical SciencesUniversity of British Columbia Vancouver British Columbia Canada
- BC Children's Hospital Research Institute Vancouver British Columbia Canada
| | - Galen E.B. Wright
- BC Children's Hospital Research Institute Vancouver British Columbia Canada
- Division of Translational TherapeuticsDepartment of PediatricsUniversity of British Columbia Vancouver British Columbia Canada
| | - Cody Lo
- BC Children's Hospital Research Institute Vancouver British Columbia Canada
- Faculty of MedicineUniversity of British Columbia Vancouver British Columbia Canada
| | - Tan Le
- Faculty of Pharmaceutical SciencesUniversity of British Columbia Vancouver British Columbia Canada
| | - Beth Brooks
- Audiology and Speech Pathology DepartmentBC Children's Hospital Vancouver British Columbia Canada
| | - Amit P. Bhavsar
- Department of Medical Microbiology and ImmunologyFaculty of Medicine and DentistryUniversity of Alberta Edmonton Alberta Canada
| | - Shahrad R. Rassekh
- BC Children's Hospital Research Institute Vancouver British Columbia Canada
- Division of Translational TherapeuticsDepartment of PediatricsUniversity of British Columbia Vancouver British Columbia Canada
| | - Colin J.D. Ross
- Faculty of Pharmaceutical SciencesUniversity of British Columbia Vancouver British Columbia Canada
- BC Children's Hospital Research Institute Vancouver British Columbia Canada
| | - Bruce C. Carleton
- BC Children's Hospital Research Institute Vancouver British Columbia Canada
- Division of Translational TherapeuticsDepartment of PediatricsUniversity of British Columbia Vancouver British Columbia Canada
| |
Collapse
|