1
|
Orasan A, Negru MC, Morgovan AI, Fleser RC, Sandu D, Sitaru AM, Motofelea AC, Balica NC. Strategies to Mitigate Cisplatin-Induced Ototoxicity: A Literature Review of Protective Agents, Mechanisms, and Clinical Gaps. Audiol Res 2025; 15:22. [PMID: 40126270 PMCID: PMC11932224 DOI: 10.3390/audiolres15020022] [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: 01/04/2025] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Cisplatin, a widely used chemotherapeutic agent, is associated with significant ototoxicity, leading to progressive and irreversible sensorineural hearing loss in up to 93% of patients. Cisplatin generates reactive oxygen species (ROS) in the cochlea, activating apoptotic and necroptotic pathways that result in hair cell death. Inflammatory processes and nitrative stress also contribute to cochlear damage. METHODS This literature review was conducted to explore the mechanisms underlying cisplatin-induced ototoxicity and evaluate protective strategies, including both current and emerging approaches. A structured search was performed in multiple scientific databases, including PubMed and ScienceDirect, for articles published up to November 2024. RESULTS Current otoprotective strategies include systemic interventions such as antioxidants, anti-inflammatory agents, and apoptosis inhibitors, as well as localized delivery methods like intratympanic injection and nanoparticle-based systems. However, these approaches have limitations, including potential interference with cisplatin's antitumor efficacy and systemic side effects. Emerging strategies focus on genetic and biomarker-based risk stratification, novel otoprotective agents targeting alternative pathways, and combination therapies. Repurposed drugs like pravastatin also show promise in reducing cisplatin-induced ototoxicity. CONCLUSIONS Despite these advancements, significant research gaps remain in translating preclinical findings to clinical applications and developing selective otoprotective agents that do not compromise cisplatin's efficacy. This review examines the mechanisms of cisplatin-induced ototoxicity, current otoprotective strategies, and emerging approaches to mitigate this adverse effect.
Collapse
Affiliation(s)
- Alexandru Orasan
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.O.); (A.I.M.); (N.C.B.)
| | - Mihaela-Cristina Negru
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.O.); (A.I.M.); (N.C.B.)
| | - Anda Ioana Morgovan
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.O.); (A.I.M.); (N.C.B.)
| | - Razvan Claudiu Fleser
- Otorhinolaryngology Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania;
| | - Daniela Sandu
- OncoHelp Cancer Centre, Radiation Oncology Department, “Victor Babes” University of Medicine and Pharmacy, Str. Rusu Sireanu nr. 34 Timisoara, 300041 Timisoara, Romania;
| | - Adrian Mihail Sitaru
- Department of Pediatric Surgery, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania;
| | - Alexandru-Catalin Motofelea
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Nicolae Constantin Balica
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.O.); (A.I.M.); (N.C.B.)
| |
Collapse
|
2
|
Arab A, Kashani B, Cordova-Delgado M, Scott EN, Alemi K, Trueman J, Groeneweg G, Chang WC, Loucks CM, Ross CJD, Carleton BC, Ester M. Machine learning model identifies genetic predictors of cisplatin-induced ototoxicity in CERS6 and TLR4. Comput Biol Med 2024; 183:109324. [PMID: 39488053 DOI: 10.1016/j.compbiomed.2024.109324] [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: 04/30/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Cisplatin-induced ototoxicity remains a significant concern in pediatric cancer treatment due to its permanent impact on quality of life. Previously, genetic association analyses have been performed to detect genetic variants associated with this adverse reaction. METHODS In this study, a combination of interpretable neural networks and Generative Adversarial Networks (GANs) was employed to identify genetic markers associated with cisplatin-induced ototoxicity. The applied method, BRI-Net, incorporates biological domain knowledge to define the network structure and employs adversarial training to learn an unbiased representation of the data, which is robust to known confounders. Leveraging genomic data from a cohort of 362 cisplatin-treated pediatric cancer patients recruited by the CPNDS (Canadian Pharmacogenomics Network for Drug Safety), this model revealed two statistically significant single nucleotide polymorphisms to be associated with cisplatin-induced ototoxicity. RESULTS Two markers within the CERS6 (rs13022792, p-value: 3 × 10-4) and TLR4 (rs10759932, p-value: 7 × 10-4) genes were associated with this cisplatin-induced adverse reaction. CERS6, a ceramide synthase, contributes to elevated ceramide levels, a known initiator of apoptotic signals in mouse models of inner ear hair cells. TLR4, a pattern-recognition protein, initiates inflammation in response to cisplatin, and reduced TLR4 expression has been shown in murine hair cells to confer protection from ototoxicity. CONCLUSION Overall, these findings provide a foundation for understanding the genetic landscape of cisplatin-induced ototoxicity, with implications for improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Ali Arab
- School of Computing Science, Simon Fraser University, Burnaby, BC, Canada
| | - Bahareh Kashani
- Department of Experimental Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | | | - Erika N Scott
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kaveh Alemi
- School of Computing Science, Simon Fraser University, Burnaby, BC, Canada
| | - Jessica Trueman
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gabriella Groeneweg
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada
| | - Wan-Chun Chang
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Catrina M Loucks
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada.
| | - Martin Ester
- School of Computing Science, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
3
|
Miao DNR, Wilke MAP, Pham J, Ladha F, Singh M, Arsenio J, Luca E, Dabdoub A, Yang W, Yang JJ, Drögemöller BI. Leveraging large-scale datasets and single cell omics data to develop a polygenic score for cisplatin-induced ototoxicity. Hum Genomics 2024; 18:112. [PMID: 39380081 PMCID: PMC11463131 DOI: 10.1186/s40246-024-00679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Cisplatin-induced ototoxicity (CIO), characterized by irreversible and progressive bilateral hearing loss, is a prevalent adverse effect of cisplatin chemotherapy. Alongside clinical risk factors, genetic variants contribute to CIO and genome-wide association studies (GWAS) have highlighted the polygenicity of this adverse drug reaction. Polygenic scores (PGS), which integrate information from multiple genetic variants across the genome, offer a promising tool for the identification of individuals who are at higher risk for CIO. Integrating large-scale hearing loss GWAS data with single cell omics data holds potential to overcome limitations related to small sample sizes associated with CIO studies, enabling the creation of PGSs to predict CIO risk. RESULTS We utilized a large-scale hearing loss GWAS and murine inner ear single nuclei RNA-sequencing (snRNA-seq) data to develop two polygenic scores: a hearing loss PGS (PGSHL) and a biologically informed PGS for CIO (PGSCIO). The PGSCIO included only variants which mapped to genes that were differentially expressed within cochlear cells that showed differential abundance in the murine snRNA-seq data post-cisplatin treatment. Evaluation of the association of these PGSs with CIO in our target CIO cohort revealed that PGSCIO demonstrated superior performance (P = 5.54 × 10- 5) relative to PGSHL (P = 2.93 × 10- 3). PGSCIO was also associated with CIO in our test cohort (P = 0.04), while the PGSHL did not show a significant association with CIO (P = 0.52). CONCLUSION This study developed the first PGS for CIO using a large-scale hearing loss dataset and a biologically informed filter generated from cisplatin-treated murine inner ear snRNA-seq data. This innovative approach offers new avenues for developing PGSs for pharmacogenomic traits, which could contribute to the implementation of tailored therapeutic interventions. Further, our approach facilitated the identification of specific cochlear cells that may play critical roles in CIO. These novel insights will guide future research aimed at developing targeted therapeutic strategies to prevent CIO.
Collapse
Affiliation(s)
- Deanne Nixie R Miao
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - MacKenzie A P Wilke
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - John Pham
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Feryal Ladha
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mansumeet Singh
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Janilyn Arsenio
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Emilia Luca
- Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Wejian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- CancerCare Manitoba Research Institute, Winnipeg, MB, Canada.
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Centre of Aging, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
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
|
Wang Y, Liu Y, Xie Y, Luan J, Liu R, Zhu Y, Ma Y, Fan Y, Sun Y, Shang W, Han F. Loss of Fascin2 increases susceptibility to cisplatin-induced hearing impairment and cochlear cell apoptosis in mice. J Otol 2024; 19:133-139. [PMID: 39735238 PMCID: PMC11681795 DOI: 10.1016/j.joto.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 06/06/2024] [Accepted: 07/02/2024] [Indexed: 12/31/2024] Open
Abstract
Objectives Deletion of Fscn2 gene in mice has been linked to progressive hearing loss and degeneration of cochlear cells. Cisplatin, an antitumor drug, can cause various side effects, including ototoxicity. The aim of this study was to investigate the effects of Fscn2 on cisplatin-induced hearing impairment in mice and to explore the possible mechanism. Methods Two-week-old Fscn2 +/+ mice and Fscn2 -/- mice were treated with two doses of cisplatin, with a 3-day recovery period in between. ABR (auditory evoked brain stem response) thresholds were measured and cochlear pathology was observed at 3 weeks of age. Results Both Fscn2 +/+ and Fscn2 -/- mice showed hearing loss under the effect of cisplatin, but the impairment was more severe in Fscn2 -/- mice. Further experiments showed that the percentages of outer hair cell (OHC) and spiral ganglion neuron (SGN) loss were significantly higher in cisplatin-treated Fscn2 -/- mice compared to Fscn2 +/+ mice. Additionally, knockdown of Fscn2 in HEI-OC1 cells worsened cisplatin-induced cell apoptosis. Conclusion FSCN2 mediates reduction of CDDP induced ototoxicity by inhibiting cell apoptosis.
Collapse
Affiliation(s)
- Yan Wang
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Yingying Liu
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Yi Xie
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Jun Luan
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Rongrong Liu
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Yongjia Zhu
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Ying Ma
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Yi Fan
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Yan Sun
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, PR China
| | - Wenjing Shang
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| | - Fengchan Han
- Department of Biochemistry and Molecular Biology, and Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, Shandong, PR China
| |
Collapse
|
6
|
Reynard P, Thai-Van H. Drug-induced hearing loss: Listening to the latest advances. Therapie 2024; 79:283-295. [PMID: 37957052 DOI: 10.1016/j.therap.2023.10.011] [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: 08/25/2023] [Accepted: 09/14/2023] [Indexed: 11/15/2023]
Abstract
Sensorineural hearing loss (SNHL) is the most common type of hearing loss. Causes include degenerative changes in the sensory hair cells, their synapses and/or the cochlear nerve. As human inner ear hair cells have no capacity for regeneration, their destruction is irreversible and leads to permanent hearing loss. SNHL can be genetically inherited or acquired through ageing, exposure to noise or ototoxic drugs. Ototoxicity generally refers to damage to the structures and functions of the inner ear following exposure to specific drugs. Ototoxicity can be multifactorial, causing damage to cochlear hair cells or cells with homeostatic functions that modulate cochlear hair cell function. Clinical strategies to limit ototoxicity include identifying patients at risk, monitoring drug concentrations, performing serial hearing assessments and switching to less ototoxic therapy. This review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, using the PubMed® database. The search terms "ototoxicity", "hearing loss" and "drugs" were combined. We included studies published between September 2013 and June 2023, and focused on medicines and drugs used in hospitals. The review highlighted a number of articles reporting the main drug classes potentially involved: namely, immunosuppressants, antimalarials, vaccines, antibiotics, antineoplastic agents, diuretics, nonsteroidal anti-inflammatory drugs and analgesics. The presumed ototoxic mechanisms were described, together with the therapeutic and preventive options developed over the last ten years.
Collapse
Affiliation(s)
- Pierre Reynard
- Service d'audiologie & explorations oto-neurologiques, hospices civils de Lyon, hôpital Edouard-Herriot & hôpital Femme Mère-Enfant, 69000 Lyon, France; Institut Pasteur, Institut de l'Audition, Center for Research and Innovation in Human Audiology, 75000 Paris, France; Université Claude Bernard Lyon 1, 69622 Villeurbanne, France
| | - Hung Thai-Van
- Service d'audiologie & explorations oto-neurologiques, hospices civils de Lyon, hôpital Edouard-Herriot & hôpital Femme Mère-Enfant, 69000 Lyon, France; Institut Pasteur, Institut de l'Audition, Center for Research and Innovation in Human Audiology, 75000 Paris, France; Université Claude Bernard Lyon 1, 69622 Villeurbanne, France.
| |
Collapse
|
7
|
Zhao YQ, Zhang HH, Wu J, Li L, Li J, Zhong H, Jin Y, Lei TY, Zhao XY, Xu B, Song QB, He J. Prediction of Tumor Microenvironment Characteristics and Treatment Response in Lung Squamous Cell Carcinoma by Pseudogene OR7E47P-related Immune Genes. Curr Med Sci 2023; 43:1133-1150. [PMID: 38015361 DOI: 10.1007/s11596-023-2798-2] [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: 04/27/2023] [Accepted: 09/22/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVE Pseudogenes are initially regarded as nonfunctional genomic sequences, but some pseudogenes regulate tumor initiation and progression by interacting with other genes to modulate their transcriptional activities. Olfactory receptor family 7 subfamily E member 47 pseudogene (OR7E47P) is expressed broadly in lung tissues and has been identified as a positive regulator in the tumor microenvironment (TME) of lung adenocarcinoma (LUAD). This study aimed to elucidate the correlation between OR7E47P and tumor immunity in lung squamous cell carcinoma (LUSC). METHODS Clinical and molecular information from The Cancer Genome Atlas (TCGA) LUSC cohort was used to identify OR7E47P-related immune genes (ORIGs) by weighted gene correlation network analysis (WGCNA). Based on the ORIGs, 2 OR7E47P clusters were identified using non-negative matrix factorization (NMF) clustering, and the stability of the clustering was tested by an extreme gradient boosting classifier (XGBoost). LASSO-Cox and stepwise regressions were applied to further select prognostic ORIGs and to construct a predictive model (ORPScore) for immunotherapy. The Botling cohorts and 8 immunotherapy cohorts (the Samstein, Braun, Jung, Gide, IMvigor210, Lauss, Van Allen, and Cho cohorts) were included as independent validation cohorts. RESULTS OR7E47P expression was positively correlated with immune cell infiltration and enrichment of immune-related pathways in LUSC. A total of 57 ORIGs were identified to classify the patients into 2 OR7E47P clusters (Cluster 1 and Cluster 2) with distinct immune, mutation, and stromal programs. Compared to Cluster 1, Cluster 2 had more infiltration by immune and stromal cells, lower mutation rates of driver genes, and higher expression of immune-related proteins. The clustering performed well in the internal and 5 external validation cohorts. Based on the 7 ORIGs (HOPX, STX2, WFS, DUSP22, SLFN13, GGCT, and CCSER2), the ORPScore was constructed to predict the prognosis and the treatment response. In addition, the ORPScore was a better prognostic factor and correlated positively with the immunotherapeutic response in cancer patients. The area under the curve values ranged from 0.584 to 0.805 in the 6 independent immunotherapy cohorts. CONCLUSION Our study suggests a significant correlation between OR7E47P and TME modulation in LUSC. ORIGs can be applied to molecularly stratify patients, and the ORPScore may serve as a biomarker for clinical decision-making regarding individualized prognostication and immunotherapy.
Collapse
Affiliation(s)
- Ya-Qi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Medical Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Hao-Han Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hao Zhong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan Jin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tian-Yu Lei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin-Yi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qi-Bin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jie He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100032, China.
| |
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
|
Garcia SL, Lauritsen J, Christiansen BK, Hansen IF, Bandak M, Dalgaard MD, Daugaard G, Gupta R. Predicting Hearing Loss in Testicular Cancer Patients after Cisplatin-Based Chemotherapy. Cancers (Basel) 2023; 15:3923. [PMID: 37568739 PMCID: PMC10417151 DOI: 10.3390/cancers15153923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Testicular cancer is predominantly curable, but the long-term side effects of chemotherapy have a severe impact on life quality. In this research study, we focus on hearing loss as a part of overall chemotherapy-induced ototoxicity. This is a unique approach where we combine clinical data from the acclaimed nationwide Danish Testicular Cancer (DaTeCa)-Late database. Clinical and genetic data on 433 patients were collected from hospital files in October 2014. Hearing loss was classified according to the FACT/GOG-Ntx-11 version 4 self-reported Ntx6. Machine learning models combining a genome-wide association study within a nested cross-validated logistic regression were applied to identify patients at high risk of hearing loss. The model comprising clinical and genetic data identified 67% of the patients with hearing loss; however, this was with a false discovery rate of 49%. For the non-affected patients, the model identified 66% of the patients with a false omission rate of 19%. An area under the receiver operating characteristic (ROC-AUC) curve of 0.73 (95% CI, 0.71-0.74) was obtained, and the model suggests genes SOD2 and MGST3 as important in improving prediction over the clinical-only model with a ROC-AUC of 0.66 (95% CI, 0.65-0.66). Such prediction models may be used to allow earlier detection and prevention of hearing loss. We suggest a possible biological mechanism for cisplatin-induced hearing loss development. On confirmation in larger studies, such models can help balance treatment in clinical practice.
Collapse
Affiliation(s)
- Sara L. Garcia
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 1958 Copenhagen, Denmark
| | - Jakob Lauritsen
- Department of Oncology, Copenhagen University Hospital, 2730 Copenhagen, Denmark
| | - Bernadette K. Christiansen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
| | - Ida F. Hansen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
| | - Mikkel Bandak
- Department of Oncology, Copenhagen University Hospital, 2730 Copenhagen, Denmark
| | - Marlene D. Dalgaard
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, 2730 Copenhagen, Denmark
| | - Ramneek Gupta
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
- Department of Computational Biology, Novo Nordisk Research Centre Oxford, Oxford OX3 7FZ, UK
| |
Collapse
|
10
|
He YQ, Luo LT, Wang TM, Xue WQ, Yang DW, Li DH, Diao H, Xiao RW, Deng CM, Zhang WL, Liao Y, Wu YX, Wang QL, Zhou T, Li XZ, Zheng XH, Zhang PF, Zhang SD, Hu YZ, Sun Y, Jia WH. Clinical and genome-wide association analysis of chemoradiation-induced hearing loss in nasopharyngeal carcinoma. Hum Genet 2023; 142:759-772. [PMID: 37062025 PMCID: PMC10182145 DOI: 10.1007/s00439-023-02554-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Chemoradiation-induced hearing loss (CRIHL) is one of the most devasting side effects for nasopharyngeal carcinoma (NPC) patients, which seriously affects survivors' long-term quality of life. However, few studies have comprehensively characterized the risk factors for CRIHL. In this study, we found that age at diagnosis, tumor stage, and concurrent cisplatin dose were positively associated with chemoradiation-induced hearing loss. We performed a genome-wide association study (GWAS) in 777 NPC patients and identified rs1050851 (within the exon 2 of NFKBIA), a variant with a high deleteriousness score, to be significantly associated with hearing loss risk (HR = 5.46, 95% CI 2.93-10.18, P = 9.51 × 10-08). The risk genotype of rs1050851 was associated with higher NFKBIA expression, which was correlated with lower cellular tolerance to cisplatin. According to permutation-based enrichment analysis, the variants mapping to 149 hereditary deafness genes were significantly enriched among GWAS top signals, which indicated the genetic similarity between hereditary deafness and CRIHL. Pathway analysis suggested that synaptic signaling was involved in the development of CRIHL. Additionally, the risk score integrating genetic and clinical factors can predict the risk of hearing loss with a relatively good performance in the test set. Collectively, this study shed new light on the etiology of chemoradiation-induced hearing loss, which facilitates high-risk individuals' identification for personalized prevention and treatment.
Collapse
Affiliation(s)
- Yong-Qiao He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Lu-Ting Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Tong-Min Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Wen-Qiong Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Da-Wei Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dan-Hua Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Hua Diao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ruo-Wen Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Chang-Mi Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Wen-Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Ying Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Yan-Xia Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Qiao-Ling Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ting Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xi-Zhao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xiao-Hui Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China
| | - Pei-Fen Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China
| | - Shao-Dan Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China
| | - Ye-Zhu Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China
| | - Ying Sun
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Wei-Hua Jia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China.
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China.
- Biobank of Sun Yat‑sen University Cancer Center, Guangzhou, People's Republic of China.
| |
Collapse
|
11
|
Association of Clinical Aspects and Genetic Variants with the Severity of Cisplatin-Induced Ototoxicity in Head and Neck Squamous Cell Carcinoma: A Prospective Cohort Study. Cancers (Basel) 2023; 15:cancers15061759. [PMID: 36980643 PMCID: PMC10046479 DOI: 10.3390/cancers15061759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Cisplatin (CDDP) is a major ototoxic chemotherapy agent for head and neck squamous cell carcinoma (HNSCC) treatment. Clinicopathological features and genotypes encode different stages of CDDP metabolism, as their coexistence may influence the prevalence and severity of hearing loss. Methods: HNSCC patients under CDDP chemoradiation were prospectively provided with baseline and post-treatment audiometry. Clinicopathological features and genetic variants encoding glutathione S-transferases (GSTT1, GSTM1, GSTP1), nucleotide excision repair (XPC, XPD, XPF, ERCC1), mismatch repair (MLH1, MSH2, MSH3, EXO1), and apoptosis (P53, CASP8, CASP9, CASP3, FAS, FASL)-related proteins were analyzed regarding ototoxicity. Results: Eighty-nine patients were included, with a cumulative CDDP dose of 260 mg/m2. Moderate/severe ototoxicity occurred in 26 (29%) patients, particularly related to hearing loss at frequencies over 3000 Hertz. Race, body-mass index, and cumulative CDDP were independent risk factors. Patients with specific isolated and combined genotypes of GSTM1, GSTP1 c.313A>G, XPC c.2815A>C, XPD c.934G>A, EXO1 c.1762G>A, MSH3 c.3133A>G, FASL c.-844A>T, and P53 c.215G>C SNVs had up to 32.22 higher odds of presenting moderate/severe ototoxicity. Conclusions: Our data present, for the first time, the association of combined inherited nucleotide variants involved in CDDP efflux, DNA repair, and apoptosis with ototoxicity, which could be potential predictors in future clinical and genomic models.
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
|
Sanchez VA, Dinh PC, Rooker J, Monahan PO, Althouse SK, Fung C, Sesso HD, Einhorn LH, Dolan ME, Frisina RD, Travis LB. Prevalence and risk factors for ototoxicity after cisplatin-based chemotherapy. J Cancer Surviv 2023; 17:27-39. [PMID: 36637632 DOI: 10.1007/s11764-022-01313-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/07/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE Ototoxicity is a prominent side effect of cisplatin-based chemotherapy. There are few reports, however, estimating its prevalence in well-defined cohorts and associated risk factors. METHODS Testicular cancer (TC) survivors given first-line cisplatin-based chemotherapy completed validated questionnaires. Descriptive statistics evaluated the prevalence of ototoxicity, defined as self-reported hearing loss and/or tinnitus. We compared patients with and without tinnitus or hearing loss using Chi-square test, two-sided Fisher's exact test, or two-sided Wilcoxon rank sum test. To evaluate ototoxicity risk factors, a backward selection logistic regression procedure was performed. RESULTS Of 145 TC survivors, 74% reported ototoxicity: 68% tinnitus; 59% hearing loss; and 52% reported both. TC survivors with tinnitus were more likely to indicate hypercholesterolemia (P = 0.008), and difficulty hearing (P < .001). Tinnitus was also significantly related to age at survey completion (OR = 1.79; P = 0.003) and cumulative cisplatin dose (OR = 5.17; P < 0.001). TC survivors with hearing loss were more likely to report diabetes (P = 0.042), hypertension (P = 0.007), hypercholesterolemia (P < 0.001), and family history of hearing loss (P = 0.044). Risk factors for hearing loss included age at survey completion (OR = 1.57; P = 0.036), hypercholesterolemia (OR = 3.45; P = 0.007), cumulative cisplatin dose (OR = 1.94; P = 0.049), and family history of hearing loss (OR = 2.87; P = 0.071). CONCLUSIONS Ototoxicity risk factors included age, cisplatin dose, cardiovascular risk factors, and family history of hearing loss. Three of four TC survivors report some type of ototoxicity; thus, follow-up of cisplatin-treated survivors should include routine assessment for ototoxicity with provision of indicated treatments. IMPLICATIONS FOR CANCER SURVIVORS Survivors should be aware of risk factors associated with ototoxicity. Referrals to audiologists before, during, and after cisplatin treatment is recommended.
Collapse
Affiliation(s)
- Victoria A Sanchez
- Department of Otolaryngology-Head & Neck Surgery, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 73, Tampa, FL, 33612, USA.
| | - Paul C Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, IN, USA
| | - Jennessa Rooker
- College of Nursing, University of South Florida, Tampa, FL, USA
| | - Patrick O Monahan
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - Sandra K Althouse
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Howard D Sesso
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lawrence H Einhorn
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Robert D Frisina
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - Lois B Travis
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
14
|
Shahbazi M, Zhang X, Dinh PC, Sanchez VA, Trendowski MR, Shuey MM, Nguyen T, Feldman DR, Vaughn DJ, Fung C, Kollmannsberger C, Martin NE, Einhorn LH, Cox NJ, Frisina RD, Travis LB, Dolan ME. Comprehensive association analysis of speech recognition thresholds after cisplatin-based chemotherapy in survivors of adult-onset cancer. Cancer Med 2023; 12:2999-3012. [PMID: 36097363 PMCID: PMC9939144 DOI: 10.1002/cam4.5218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/05/2022] Open
Abstract
PURPOSE Deficits in speech understanding constitute one of the most severe consequences of hearing loss. Here we investigate the clinical and genetic risk factors for symmetric deterioration of speech recognition thresholds (SRT) among cancer survivors treated with cisplatin. METHODS SRT was measured using spondaic words and calculating the mean of measurements for both ears with symmetric SRT values. For clinical associations, SRT-based hearing disability (SHD) was defined as SRT≥15 dB hearing loss and clinical variables were derived from the study dataset. Genotyped blood samples were used for GWAS with rank-based inverse normal transformed SRT values as the response variable. Age was used as a covariate in association analyses. RESULTS SHD was inversely associated with self-reported health (p = 0.004). Current smoking (p = 0.002), years of smoking (p = 0.02), BMI (p < 0.001), and peripheral motor neuropathy (p = 0.003) were positively associated with SHD, while physical activity was inversely associated with SHD (p = 0.005). In contrast, cumulative cisplatin dose, peripheral sensory neuropathy, hypertension, and hypercholesterolemia were not associated with SHD. Although no genetic variants had an association p value < 5 × 10-8 , 22 genetic variants were suggestively associated (p < 10-5 ) with SRT deterioration. Three of the top variants in 10 respective linkage disequilibrium regions were either positioned within the coding sequence or were eQTLs for genes involved in neuronal development (ATE1, ENAH, and ZFHX3). CONCLUSION Current results improve our understanding of risk factors for SRT deterioration in cancer survivors. Higher BMI, lower physical activity, and smoking are associated with SHD. Larger samples would allow for expansion of the current findings on the genetic architecture of SRT.
Collapse
Affiliation(s)
| | - Xindi Zhang
- Department of MedicineUniversity of ChicagoChicagoIllinoisUSA
| | - Paul C. Dinh
- Department of Medical OncologyIndiana UniversityIndianapolisIndianaUSA
| | - Victoria A. Sanchez
- Department of Otolaryngology—Head and Neck SurgeryUniversity of South FloridaTampaFloridaUSA
| | | | - Megan M. Shuey
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tessa Nguyen
- Center for Audiology, Speech, Language and LearningNorthwesthern UniversityChicagoIllinoisUSA
| | | | - Darren R. Feldman
- Department of Medical Oncology, Memorial Sloan‐Kettering Cancer CenterNew YorkNew YorkUSA
| | - David J. Vaughn
- Department of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical CenterRochesterNew YorkUSA
| | | | - Neil E. Martin
- Department of Radiation OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | | | - Nancy J. Cox
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Robert D. Frisina
- Departments of Medical Engineering and Communication Sciences and Disorders, Global Center for Hearing and Speech ResearchUniversity of South FloridaTampaFloridaUSA
| | - Lois B. Travis
- Department of Medical OncologyIndiana UniversityIndianapolisIndianaUSA
| | | |
Collapse
|
15
|
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: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
16
|
Hurkmans EGE, Klumpers MJ, Dello Russo C, De Witte W, Guchelaar HJ, Gelderblom H, Cleton-Jansen AM, Vermeulen SH, Kaal S, van der Graaf WTA, Flucke U, Gidding CEM, Schreuder HWB, de Bont ESJM, Caron HN, Gattuso G, Schiavello E, Terenziani M, Massimino M, McCowage G, Nagabushan S, Limaye A, Rose V, Catchpoole D, Jorgensen AL, Barton C, Delaney L, Hawcutt DB, Pirmohamed M, Pizer B, Coenen MJH, te Loo DMWM. Genome-wide analyses of platinum-induced ototoxicity in childhood cancer patients: Results of GO-CAT and United Kingdom MAGIC consortia. Front Pharmacol 2023; 13:980309. [PMID: 36699085 PMCID: PMC9870026 DOI: 10.3389/fphar.2022.980309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023] Open
Abstract
Hearing loss (ototoxicity) is a major adverse effect of cisplatin and carboplatin chemotherapy. The aim of this study is to identify novel genetic variants that play a role in platinum-induced ototoxicity. Therefore, a genome-wide association study was performed in the Genetics of Childhood Cancer Treatment (GO-CAT) cohort (n = 261) and the United Kingdom Molecular Genetics of Adverse Drug Reactions in Children Study (United Kingdom MAGIC) cohort (n = 248). Results of both cohorts were combined in a meta-analysis. In primary analysis, patients with SIOP Boston Ototoxicity Scale grade ≥1 were considered cases, and patients with grade 0 were controls. Variants with a p-value <10-5 were replicated in previously published data by the PanCareLIFE cohort (n = 390). No genome-wide significant associations were found, but variants in TSPAN5, RBBP4P5, AC010090.1 and RNU6-38P were suggestively associated with platinum-induced ototoxicity. The lowest p-value was found for rs7671702 in TSPAN5 (odds ratio 2.0 (95% confidence interval 1.5-2.7), p-value 5.0 × 10-7). None of the associations were significant in the replication cohort, although the effect directions were consistent among all cohorts. Validation and functional understanding of these genetic variants could lead to more insights in the development of platinum-induced ototoxicity.
Collapse
Affiliation(s)
| | - Marije J. Klumpers
- Department of Pediatrics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cinzia Dello Russo
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom,Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ward De Witte
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Sita H. Vermeulen
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, Netherlands
| | - Suzanne Kaal
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Winette T. A. van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands,Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Uta Flucke
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | - Eveline S. J. M. de Bont
- Department of Pediatrics, Beatrix Children’s Hospital, University Medical Center Groningen, Groningen, Netherlands
| | - Huib N. Caron
- Department of Pediatrics, Amsterdam University Medical Centers, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Giovanna Gattuso
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisabetta Schiavello
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Terenziani
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Geoff McCowage
- Cancer Centre for Children, The Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Sumanth Nagabushan
- Cancer Centre for Children, The Children’s Hospital at Westmead, Sydney, NSW, Australia,Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW, Australia
| | - Anuja Limaye
- Department of Audiology, The Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Victoria Rose
- Department of Neuro-Otology, Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia
| | - Daniel Catchpoole
- Children’s Cancer Research Unit, The Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Andrea L. Jorgensen
- Department of Health Data Science, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Barton
- Department of Women’s and Children’s Health, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Delaney
- Department of Women’s and Children’s Health, University of Liverpool, Liverpool, United Kingdom
| | - Daniel B. Hawcutt
- Department of Women’s and Children’s Health, University of Liverpool, Liverpool, United Kingdom,NIHR Alder Hey Clinical Research Facility, Alder Hey Children’s Hospital, Liverpool, United Kingdom
| | - Munir Pirmohamed
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Barry Pizer
- Department of Pediatric Oncology, Alder Hey Children’s Hospital, Liverpool, United Kingdom
| | - Marieke J. H. Coenen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - D. Maroeska W. M. te Loo
- Department of Pediatrics, Radboud University Medical Center, Nijmegen, Netherlands,*Correspondence: D. Maroeska W. M. te Loo,
| |
Collapse
|
17
|
Reizine N, O’Donnell PH. Modern developments in germline pharmacogenomics for oncology prescribing. CA Cancer J Clin 2022; 72:315-332. [PMID: 35302652 PMCID: PMC9262778 DOI: 10.3322/caac.21722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
The integration of genomic data into personalized treatment planning has revolutionized oncology care. Despite this, patients with cancer remain vulnerable to high rates of adverse drug events and medication inefficacy, affecting prognosis and quality of life. Pharmacogenomics is a field seeking to identify germline genetic variants that contribute to an individual's unique drug response. Although there is widespread integration of genomic information in oncology, somatic platforms, rather than germline biomarkers, have dominated the attention of cancer providers. Patients with cancer potentially stand to benefit from improved integration of both somatic and germline genomic information, especially because the latter may complement treatment planning by informing toxicity risk for drugs with treatment-limiting tolerabilities and narrow therapeutic indices. Although certain germline pharmacogenes, such as TPMT, UGT1A1, and DPYD, have been recognized for decades, recent attention has illuminated modern potential dosing implications for a whole new set of anticancer agents, including targeted therapies and antibody-drug conjugates, as well as the discovery of additional genetic variants and newly relevant pharmacogenes. Some of this information has risen to the level of directing clinical action, with US Food and Drug Administration label guidance and recommendations by international societies and governing bodies. This review is focused on key new pharmacogenomic evidence and oncology-specific dosing recommendations. Personalized oncology care through integrated pharmacogenomics represents a unique multidisciplinary collaboration between oncologists, laboratory science, bioinformatics, pharmacists, clinical pharmacologists, and genetic counselors, among others. The authors posit that expanded consideration of germline genetic information can further transform the safe and effective practice of oncology in 2022 and beyond.
Collapse
Affiliation(s)
- Natalie Reizine
- Division of Hematology and Oncology, Department of Medicine, The University of Illinois at Chicago
| | - Peter H. O’Donnell
- Section of Hematology/Oncology, Department of Medicine, Center for Personalized Therapeutics, and Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago
- Correspondence to: Dr. Peter H. O’Donnell, Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA. ()
| |
Collapse
|
18
|
Zou X, Wang R, Yang Z, Wang Q, Fu W, Huo Z, Ge F, Zhong R, Jiang Y, Li J, Xiong S, Hong W, Liang W. Family Socioeconomic Position and Lung Cancer Risk: A Meta-Analysis and a Mendelian Randomization Study. Front Public Health 2022; 10:780538. [PMID: 35734761 PMCID: PMC9207765 DOI: 10.3389/fpubh.2022.780538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundFamily socioeconomic position (SEP) in childhood is an important factor to predict some chronic diseases. However, the association between family SEP in childhood and the risk of lung cancer is not clear.MethodsA systematic search was performed to explore their relationship. We selected education level, socioeconomic positions of parents and childhood housing conditions to represent an individual family SEP. Hazard ratios (HRs) of lung cancer specific-mortality were synthesized using a random effects model. Two-sample Mendelian randomization (MR) was carried out with summary data from published genome-wide association studies of SEP to assess the possible causal relationship of SEP and risk of lung cancer.ResultsThrough meta-analysis of 13 studies, we observed that to compared with the better SEP, the poorer SEP in the childhood was associated with the increased lung cancer risk in the adulthood (HR: 1.25, 95% CI: 1.10 to 1.43). In addition, the dose-response analysis revealed a positive correlation between the poorer SEP and increased lung cancer risk. Same conclusion was reached in MR [(education level) OR 0.50, 95% CI: 0.39 to 0.63; P < 0.001].ConclusionThis study indicates that poor family socioeconomic position in childhood is causally correlated with lung cancer risk in adulthood.Systematic Review Registrationidentifier: 159082.
Collapse
Affiliation(s)
- Xusen Zou
- South China University of Technology, School of Public Administration, Guangzhou, China
| | - Runchen Wang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Zhao Yang
- Peking University First Hospital, Beijing, China
| | - Qixia Wang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Wenhai Fu
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- First Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Zhenyu Huo
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Fan Ge
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- First Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Ran Zhong
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Jiang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Jiangfu Li
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shan Xiong
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wen Hong
- South China University of Technology, School of Public Administration, Guangzhou, China
- Wen Hong
| | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Wenhua Liang
| |
Collapse
|
19
|
Wibmer AG, Dinh PC, Travis LB, Chen C, Bromberg M, Zheng J, Capanu M, Sesso HD, Feldman DR, Vargas HA. Associations of Body Fat Distribution and Cardiometabolic Risk of Testicular Cancer Survivors after Cisplatin-Based Chemotherapy. JNCI Cancer Spectr 2022; 6:6585341. [PMID: 35801305 PMCID: PMC9263534 DOI: 10.1093/jncics/pkac030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
Background
It is unknown how body fat distribution modulates the cardiometabolic risk of testicular cancer survivors (TCSs) after cisplatin-based chemotherapy.
Methods
For 455 patients enrolled in The Platinum Study at Memorial Sloan Kettering Cancer Center, visceral (VAT) and subcutaneous (SAT) adipose tissue was quantified on pre-chemotherapy CT. VAT/SAT ratio was calculated as a quantitative measure of central adiposity. Endpoints were incidence of new post-chemotherapy cardiometabolic disease (new antihypertensive, lipid-lowering, or diabetes medication), and post-chemotherapy Framingham risk scores. Cox models and linear regression with interaction terms were applied. Post-chemotherapy body fat distribution was analyzed in 108 patients. All statistical tests were 2-sided.
Results
Baseline median age was 31 years (IQR = 26, 39), BMI 26 kg/m2 (IQR: 24, 29), and VAT/SAT ratio 0.49 (IQR: 0.31, 0.75). Median follow-up was 26 months (IQR: 16, 59). Higher pre-chemotherapy VAT/SAT ratios inferred a higher likelihood of new cardiometabolic disease among patients with BMI ≥30 kg/m2 (age-adjusted HR = 3.14, 95% CI = 1.02–9.71, p = 0.047), but not other BMI groups. Pre-chemotherapy VAT/SAT ratio was associated with post-chemotherapy Framingham risk scores in univariate regression analysis (exp(β)-estimate: 2.10, 95% CI: 1.84, 2.39, p < 0.001); in a multivariate model, this association was stronger in younger versus older individuals. BMI increased in most patients after chemotherapy and correlated with increases in VAT/SAT (Spearman r = 0.39; p < 0.001).
Conclusions
In TCSs, central adiposity is associated with increased cardiometabolic risk after cisplatin-based chemotherapy, particularly in obese or young men. Weight gain after chemotherapy occurs preferentially in the visceral compartment, providing insight into the pathogenesis of cardiovascular disease in this population.
Collapse
Affiliation(s)
- Andreas G Wibmer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul C Dinh
- Indiana University School of Medicine, Department of Medicine, Indianapolis, IN, USA
| | - Lois B Travis
- Indiana University School of Medicine, Department of Medicine, Indianapolis, IN, USA
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, USA
| | - Carol Chen
- Department of Medicine, Cardiology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Bromberg
- Department of Medicine, Genitourinary Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Junting Zheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Howard D Sesso
- Brigham and Women's Hospital, Department of Medicine, Boston, MA, USA
| | - Darren R Feldman
- Department of Medicine, Genitourinary Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
20
|
Zhang X, Trendowski MR, Wilkinson E, Shahbazi M, Dinh PC, Shuey MM, Feldman DR, Hamilton RJ, Vaughn DJ, Fung C, Kollmannsberger C, Huddart R, Martin NE, Sanchez VA, Frisina RD, Einhorn LH, Cox NJ, Travis LB, Dolan ME. Pharmacogenomics of cisplatin-induced neurotoxicities: Hearing loss, tinnitus, and peripheral sensory neuropathy. Cancer Med 2022; 11:2801-2816. [PMID: 35322580 PMCID: PMC9302309 DOI: 10.1002/cam4.4644] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Cisplatin is a critical component of first-line chemotherapy for several cancers, but causes peripheral sensory neuropathy, hearing loss, and tinnitus. We aimed to identify comorbidities for cisplatin-induced neurotoxicities among large numbers of similarly treated patients without the confounding effect of cranial radiotherapy. METHODS Utilizing linear and logistic regression analyses on 1680 well-characterized cisplatin-treated testicular cancer survivors, we analyzed associations of hearing loss, tinnitus, and peripheral neuropathy with nongenetic comorbidities. Genome-wide association studies and gene-based analyses were performed on each phenotype. RESULTS Hearing loss, tinnitus, and peripheral neuropathy, accounting for age and cisplatin dose, were interdependent. Survivors with these neurotoxicities experienced more hypertension and poorer self-reported health. In addition, hearing loss was positively associated with BMIs at clinical evaluation and nonwork-related noise exposure (>5 h/week). Tinnitus was positively associated with tobacco use, hypercholesterolemia, and noise exposure. We observed positive associations between peripheral neuropathy and persistent vertigo, tobacco use, and excess alcohol consumption. Hearing loss and TXNRD1, which plays a key role in redox regulation, showed borderline significance (p = 4.2 × 10-6 ) in gene-based analysis. rs62283056 in WFS1 previously found to be significantly associated with hearing loss (n = 511), was marginally significant in an independent replication cohort (p = 0.06; n = 606). Gene-based analyses identified significant associations between tinnitus and WNT8A (p = 2.5 × 10-6 ), encoding a signaling protein important in germ cell tumors. CONCLUSIONS Genetics variants in TXNRD1 and WNT8A are notable risk factors for hearing loss and tinnitus, respectively. Future studies should investigate these genes and if replicated, identify their potential impact on preventive strategies.
Collapse
Affiliation(s)
- Xindi Zhang
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | - Emma Wilkinson
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Mohammad Shahbazi
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Paul C Dinh
- Division of Medical Oncology, Indiana University, Indianapolis, Indiana, USA
| | - Megan M Shuey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Darren R Feldman
- Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Robert J Hamilton
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David J Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | - Neil E Martin
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Victoria A Sanchez
- Department of Otolaryngology - Head and Neck Surgery, University of South Florida, Tampa, Florida, USA
| | - Robert D Frisina
- Departments of Medical Engineering and Communication Sciences and Disorders, Global Center for Hearing and Speech Research, University of South Florida, Tampa, Florida, USA
| | - Lawrence H Einhorn
- Division of Medical Oncology, Indiana University, Indianapolis, Indiana, USA
| | - Nancy J Cox
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lois B Travis
- Division of Medical Oncology, Indiana University, Indianapolis, Indiana, USA.,Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
21
|
Genome-wide analysis identify novel germline genetic variations in ADCY1 influencing platinum-based chemotherapy response in non-small cell lung cancer. Acta Pharm Sin B 2022; 12:1514-1522. [PMID: 35530157 PMCID: PMC9069400 DOI: 10.1016/j.apsb.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 11/21/2022] Open
Abstract
To explore the pharmacogenomic markers that affect the platinum-based chemotherapy response in non-small-cell lung carcinoma (NSCLC), we performed a two-cohort of genome-wide association studies (GWAS), including 34 for WES-based and 433 for microarray-based analyses, as well as two independent validation cohorts. After integrating the results of two studies, the genetic variations related to the platinum-based chemotherapy response were further determined by fine-mapping in 838 samples, and their potential functional impact were investigated by eQTL analysis and in vitro cell experiments. We found that a total of 68 variations were significant at P < 1 × 10-3 in cohort 1 discovery stage, of which 3 SNPs were verified in 262 independent samples. A total of 541 SNPs were significant at P < 1 × 10-4 in cohort 2 discovery stage, of which 8 SNPs were verified in 347 independent samples. Comparing the validated SNPs in two GWAS, ADCY1 gene was verified in both independent studies. The results of fine-mapping showed that the G allele carriers of ADCY1 rs2280496 and C allele carriers of rs189178649 were more likely to be resistant to platinum-based chemotherapy. In conclusion, our study found that rs2280496 and rs189178649 in ADCY1 gene were associated the sensitivity of platinum-based chemotherapy in NSCLC patients.
Collapse
|
22
|
Finch LE, Cardonick EH. Incidence of childhood hearing loss after in utero exposure to platinum agents. Prenat Diagn 2021; 41:1467-1474. [PMID: 34462927 DOI: 10.1002/pd.6035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE When treated for childhood cancers, at least 50% of children exposed to platinum agents have permanent hearing loss. We determined the relative risk of childhood hearing loss after in utero exposure to platinum chemotherapy in our registry cohort. METHOD After exposure to platinum chemotherapy in utero, all children undergo routine newborn hearing screening. This consists of Otoacoustic Emissions. Children who failed this screen were evaluated by audiologists. For those children with hearing loss by Automated Auditory Brainstem Response, prenatal and postnatal treatment details were compared to platinum exposed children without hearing loss. RESULTS Three hundred and seven children were exposed to chemotherapy in utero. Four children were diagnosed with hearing loss, all exposed to platinum agents. Chemotherapy exposures included: Cisplatin/Paclitaxel (2), Etoposide/Cisplatin/Bleomycin (1), Carboplatin/Paclitaxel (1) to treat ovarian (2), or cervical cancer (2). Of the 39 platinum exposed without hearing loss: 11 children were exposed to oxaliplatin, 16 were exposed to cisplatin and 12 to carboplatin in utero. Two hundred and sixty four women received non-platinum based chemotherapy for various cancers during pregnancy. Among these, there were no cases of hearing loss. There was a significant difference in hearing loss based on exposure to platinum agents in utero compared to non-platinum-containing chemotherapy regimens, 4/43 versus 0/264, p = 0.0003. There were no statistical differences in prenatal and postnatal treatment details, including: gestational age at diagnosis, at first chemotherapy treatment, at first platinum treatment, at delivery (<32 weeks, <35 weeks, <37 weeks), gender, birthweight, birthweight percentile, rates of intrauterine growth restriction, neonatal complications or use of postnatal antibiotics between the platinum exposed children with and without hearing loss. CONCLUSION The only children in the registry exposed to chemotherapy who were diagnosed with hearing loss had been exposed to cisplatin or carboplatin in utero. No hearing loss occurred in children exposed to oxaliplatin, or non-platinum agents. Due to a concern for cisplatin ototoxicity, carboplatin is the preferred platinum agent for use in pregnancy when equivalent maternal survival can be expected for the particular cancer type. For newborns exposed to platinum agents in utero, newborn screening with an auditory emissions test at birth (OES) may not detect sensorineural hearing loss and auditory brainstem response testing is recommended, regardless of the newborn screening result.
Collapse
Affiliation(s)
- Lauren E Finch
- Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Elyce H Cardonick
- Maternal Fetal Medicine Department, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| |
Collapse
|
23
|
Meijer AJM, van den Heuvel-Eibrink MM, Brooks B, Am Zehnhoff-Dinnesen AG, Knight KR, Freyer DR, Chang KW, Hero B, Papadakis V, Frazier AL, Blattmann C, Windsor R, Morland B, Bouffet E, Rutkowski S, Tytgat GAM, Geller JI, Hunter LL, Sung L, Calaminus G, Carleton BC, Helleman HW, Foster JH, Kruger M, Cohn RJ, Landier W, van Grotel M, Brock PR, Hoetink AE, Rajput KM. Recommendations for Age-Appropriate Testing, Timing, and Frequency of Audiologic Monitoring During Childhood Cancer Treatment: An International Society of Paediatric Oncology Supportive Care Consensus Report. JAMA Oncol 2021; 7:1550-1558. [PMID: 34383016 DOI: 10.1001/jamaoncol.2021.2697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Ototoxicity is an irreversible direct and late effect of certain childhood cancer treatments. Audiologic surveillance during therapy as part of the supportive care pathway enables early detection of hearing loss, decision-making about ongoing cancer treatment, and, when applicable, the timely use of audiologic interventions. Pediatric oncologic clinical practice and treatment trials have tended to be driven by tumor type and tumor-specific working groups. Internationally accepted standardized recommendations for monitoring hearing during treatment have not previously been agreed on. Objective To provide standard recommendations on hearing loss monitoring during childhood cancer therapy for clinical practice. Methods An Ototoxicity Task Force was formed under the umbrella of the International Society of Paediatric Oncology, consisting of international audiologists, otolaryngologists, and leaders in the field of relevant pediatric oncology tumor groups. Consensus meetings conducted by experts were organized, aimed at providing standardized recommendations on age-directed testing, timing, and frequency of monitoring during cancer treatment based on literature and consensus. Consensus statements were prepared by the core group, adapted following several videoconferences, and finally agreed on by the expert panel. Findings The consensus reached was that children who receive ototoxic cancer treatment (platinum agents, cranial irradiation, and/or brain surgery) require a baseline case history, monitoring of their middle ear and inner ear function, and assessment of tinnitus at each audiologic follow-up. As a minimum, age-appropriate testing should be performed before and at the end of treatment. Ideally, audiometry with counseling before each cisplatin cycle should be considered in the context of the individual patient, specific disease, feasibility, and available resources. Conclusions and Relevance This is an international multidisciplinary consensus report providing standardized supportive care recommendations on hearing monitoring in children undergoing potentially ototoxic cancer treatment. The recommendations are intended to improve the care of children with cancer and facilitate comparative research on the timing and development of hearing loss caused by different cancer treatment regimens.
Collapse
Affiliation(s)
- Annelot J M Meijer
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Beth Brooks
- Department of Audiology and Speech Pathology, BC Children's Hospital, Vancouver, British Columbia, Canada.,School of Audiology and Speech Science, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Kristin R Knight
- Department of Pediatric Audiology, Oregon Health and Science University, Portland
| | - David R Freyer
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles.,Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles.,Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles
| | - Kay W Chang
- Department of Otolaryngology, Stanford University School of Medicine, Palo Alto, California
| | - Barbara Hero
- Department of Pediatric Hematology-Oncology, Children's Hospital, University of Cologne, Cologne, Germany
| | - Vassilios Papadakis
- Department of Pediatric Hematology-Oncology, Agia Sofia Children's Hospital, Athens, Greece
| | - A Lindsay Frazier
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Claudia Blattmann
- Department of Pediatric Oncology/Hematology/Immunology, Stuttgart Cancer Center, Olgahospital Stuttgart, Stuttgart, Germany
| | - Rachael Windsor
- Department of Oncology, University College London Hospitals National Health Service Trust, London, United Kingdom
| | - Bruce Morland
- Department of Pediatric Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Hamburg, Germany
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Lisa L Hunter
- Communication Sciences Research Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Lillian Sung
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Program in Child Health Evaluative Sciences, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Canada
| | - Gabriele Calaminus
- Department of Pediatric Hematology and Oncology, University Hospital Bonn, Bonn, Germany
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Hiske W Helleman
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Utrecht-Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Jennifer H Foster
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Centers, Houston, Texas
| | - Mariana Kruger
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Richard J Cohn
- Kids Cancer Centre, Sydney Children's Hospital, High Street, Randwick, Australia.,School of Women's and Children's Health, University of New South Wales Medicine, Sydney, Australia
| | - Wendy Landier
- Department of Pediatrics, Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham
| | - Martine van Grotel
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Penelope R Brock
- Department of Pediatric Oncology, Great Ormond Street Hospital for Children National Health Service Trust, London, United Kingdom
| | - Alexander E Hoetink
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Utrecht-Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Kaukab M Rajput
- Department of Audiovestibular Medicine and Cochlear Implant, Great Ormond Street Hospital for Children National Health Service Trust, London, United Kingdom
| | | |
Collapse
|
24
|
King J, Adra N, Einhorn LH. Testicular cancer: Biology to bedside. Cancer Res 2021; 81:5369-5376. [PMID: 34380632 DOI: 10.1158/0008-5472.can-21-1452] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/27/2021] [Accepted: 08/10/2021] [Indexed: 11/16/2022]
Abstract
Testicular cancer is the first solid tumor with a remarkably high cure rate. This success was only made possible through collaborative efforts of basic and clinical research. Most patients with distant metastases can be cured. However, the majority of these patients are diagnosed at a young age, leaving many decades for the development of treatment-related complications. This has magnified the importance of research into survivorship issues after exposure to platinum-based chemotherapy. This research, along with research into newer biomarkers that will aid in the diagnosis and surveillance of patients and survivors of testicular cancer, will continue to advance the field and provide new opportunities for these patients. There also remains the need for further therapeutic options for patients who unfortunately do not respond to standard treatment regimens and ultimately die from this disease, including a cohort of patients with late relapses and platinum-refractory disease. Here we discuss the advancements in management that led to a highly curable malignancy, while highlighting difficult situations still left to solve as well as emerging research into novel biomarkers.
Collapse
Affiliation(s)
- Jennifer King
- Hematology/Oncology, Indiana University School of Medicine
| | - Nabil Adra
- Hematology/Oncology, Indiana University School of Medicine
| | | |
Collapse
|
25
|
Trendowski MR, Baedke JL, Sapkota Y, Travis LB, Zhang X, El Charif O, Wheeler HE, Leisenring WM, Robison LL, Hudson MM, Morton LM, Oeffinger KC, Howell RM, Armstrong GT, Bhatia S, Dolan ME. Clinical and genetic risk factors for radiation-associated ototoxicity: A report from the Childhood Cancer Survivor Study and the St. Jude Lifetime Cohort. Cancer 2021; 127:4091-4102. [PMID: 34286861 DOI: 10.1002/cncr.33775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cranial radiation therapy (CRT) is associated with ototoxicity, which manifests as hearing loss and tinnitus. The authors sought to identify clinical determinants and genetic risk factors for ototoxicity among adult survivors of pediatric cancer treated with CRT. METHODS Logistic regression evaluated associations of tinnitus (n = 1991) and hearing loss (n = 2198) with nongenetic risk factors and comorbidities among CRT-treated survivors in the Childhood Cancer Survivor Study. Genome-wide association studies (GWASs) of CRT-related tinnitus and hearing loss were also performed. RESULTS Males were more likely to report CRT-related tinnitus (9.4% vs 5.4%; P = 5.1 × 10-4 ) and hearing loss (14.0% vs 10.7%; P = .02) than females. Survivors with tinnitus or hearing loss were more likely to experience persistent dizziness or vertigo (tinnitus: P < 2 × 10-16 ; hearing loss: P = 6.4 × 10-9 ), take antidepressants (tinnitus: P = .02; hearing loss: P = .01), and report poorer overall health (tinnitus: P = 1.5 × 10-6 ; hearing loss: P = 1.7 × 10-6 ) in comparison with controls. GWAS of CRT-related tinnitus revealed a genome-wide significant signal in chromosome 1 led by rs203248 (P = 1.5 × 10-9 ), whereas GWAS of CRT-related hearing loss identified rs332013 (P = 5.8 × 10-7 ) in chromosome 8 and rs67522722 (P = 7.8 × 10-7 ) in chromosome 6 as nearly genome-wide significant. A replication analysis identified rs67522722, intronic to ATXN1, as being significantly associated with CRT-related hearing loss (P = .03) and de novo hearing loss (P = 3.6 × 10-4 ). CONCLUSIONS CRT-associated ototoxicity was associated with sex, several neuro-otological symptoms, increased antidepressant use, and poorer self-reported health. GWAS of CRT-related hearing loss identified rs67522722, which was supported in an independent cohort of survivors. LAY SUMMARY Hearing loss and subjective tinnitus (the perception of noise or ringing in the ear) are long-term side effects of cancer treatment and are common in children treated with radiation to the brain. These toxicities can affect childhood development and potentially contribute to serious learning and behavioral difficulties. This study's data indicate that males are at greater risk for hearing loss and tinnitus than females after radiation therapy to the brain. Those who develop these toxicities are more likely to use antidepressants and report poorer overall health. Health care providers can improve the management of survivors by informing patients and/or their parents of these risks.
Collapse
Affiliation(s)
| | - Jessica L Baedke
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lois B Travis
- Department of Medical Oncology, Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana.,Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana
| | - Xindi Zhang
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Omar El Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Heather E Wheeler
- Department of Biology, Loyola University Chicago, Chicago, Illinois.,Department of Computer Science, Loyola University Chicago, Chicago, Illinois
| | - Wendy M Leisenring
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lindsay M Morton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Rebecca M Howell
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Smita Bhatia
- School of Medicine Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
26
|
Chovanec M, Lauritsen J, Bandak M, Oing C, Kier GG, Kreiberg M, Rosenvilde J, Wagner T, Bokemeyer C, Daugaard G. Late adverse effects and quality of life in survivors of testicular germ cell tumour. Nat Rev Urol 2021; 18:227-245. [PMID: 33686290 DOI: 10.1038/s41585-021-00440-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Currently, ~95% of patients with testicular germ cell tumour (TGCT) are cured, resulting in an increasing number of TGCT survivors. Although cured, these men face potential late adverse effects and reduced quality of life. Survivors face a twofold increased risk of second malignant neoplasms after chemotherapy and radiotherapy, with evidence of dose-dependent associations. For survivors managed with surveillance or treated with radiotherapy, the risk of cardiovascular disease (CVD) is comparable to the risk in the general population, whereas treatment with chemotherapy increases the risk of life-threatening CVD, especially during treatment and after 10 years of follow-up. Other adverse effects are organ-related toxicities such as neuropathy and ototoxicity. Pulmonary and renal impairment in patients with TGCT treated with chemotherapy is limited. Survivors of TGCT might experience psychosocial distress including anxiety disorders, fear of cancer recurrence and TGCT-specific issues, such as sexual dysfunction. Late adverse effects can be avoided in most patients with stage I disease if followed on a surveillance programme. However, patients with disseminated disease can experience toxicities associated with radiotherapy and chemotherapy, and/or adverse effects related to surgery for residual disease. The severity of adverse effects increases with dose of both chemotherapy and radiotherapy. This Review discusses the most recent data concerning the late adverse effects of today's standard treatments for TGCT.
Collapse
Affiliation(s)
- Michal Chovanec
- 2nd Department of Oncology, Comenius University, National Cancer Institute, Bratislava, Slovakia
| | - Jakob Lauritsen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Mikkel Bandak
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christoph Oing
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gry Gundgaard Kier
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Kreiberg
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Josephine Rosenvilde
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Wagner
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
27
|
Ouellette TW, Wright GE, Drögemöller BI, Ross CJ, Carleton BC. Integrating disease and drug-related phenotypes for improved identification of pharmacogenomic variants. Pharmacogenomics 2021; 22:251-261. [PMID: 33769074 DOI: 10.2217/pgs-2020-0130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: To improve the identification and interpretation of pharmacogenetic variants through the integration of disease and drug-related traits. Materials & methods: We hypothesized that integrating genome-wide disease and pharmacogenomic data may drive new insights into drug toxicity and response by identifying shared genetic architecture. Pleiotropic variants were identified using a methodological framework incorporating colocalization analysis. Results: Using genome-wide association studies summary statistics from the UK Biobank, European Bioinformatics Institute genome-wide association studies catalog and the Pharmacogenomics Research Network, we validated pleiotropy at the ABCG2 locus between allopurinol response and gout and identified novel pleiotropy between antihypertensive-induced new-onset diabetes, Crohn's disease and inflammatory bowel disease at the IL18RAP/SLC9A4 locus. Conclusion: New mechanistic insights and genetic loci can be uncovered by identifying pleiotropy between disease and drug-related traits.
Collapse
Affiliation(s)
- Tom W Ouellette
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
| | - Galen Eb Wright
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Britt I Drögemöller
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Colin Jd Ross
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
28
|
Kim H, Choe SA, Lee SJ, Sung J. Causal relationship between the timing of menarche and young adult body mass index with consideration to a trend of consistently decreasing age at menarche. PLoS One 2021; 16:e0247757. [PMID: 33635908 PMCID: PMC7909625 DOI: 10.1371/journal.pone.0247757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Younger age at menarche (AAM) is associated with higher body mass index (BMI) for young women. Considering that continuous trends in decreasing AAM and increasing BMI are found in many countries, we attempted to assess whether the observed negative association between AAM and young adult BMI is causal. We included 4,093 women from the Korean Genome and Epidemiology Study (KoGES) and Healthy twin Study (HTS) with relevant epidemiologic data and genome-wide marker information. To mitigate the remarkable differences in AAM across generations, we converted the AAM to a generation-standardized AAM (gsAAM). To test causality, we applied the Mendelian randomization (MR) approach, using a genetic risk score (GRS) based on 14 AAM-associated single nucleotide polymorphisms (SNPs). We constructed MR models adjusting for education level and validated the results using the inverse-variance weighted (IVW), weighted median (WM), MR-pleiotropy residual sum and outliers test (MR-PRESSO), and MR-Egger regression methods. We found a null association using observed AAM and BMI level (conventional regression; -0.05 [95% CIs -0.10-0.00] per 1-year higher AAM). This null association was replicated when gsAAM was applied instead of AAM. Using the two-stage least squares (2SLS) approach employing a univariate GRS, the association was also negated for both AAM and gsAAM, regardless of model specifications. All the MR diagnostics suggested statistically insignificant associations, but weakly negative trends, without evidence of confounding from pleiotropy. We did not observe a causal association between AAM and young adult BMI whether we considered the birth cohort effect or not. Our study alone does not exclude the possibility of existing a weak negative association, considering the modest power of our study design.
Collapse
Affiliation(s)
- Hakyung Kim
- Genome and Health Big Data Laboratory, Department of Public Health, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Seung-Ah Choe
- Department of Preventive Medicine, Korea University College of Medicine, Seoul, Korea
| | - Soo Ji Lee
- Genome and Health Big Data Laboratory, Department of Public Health, Graduate School of Public Health, Seoul National University, Seoul, Korea
- Institute of Health & Environment, Seoul National University, Seoul, Korea
| | - Joohon Sung
- Genome and Health Big Data Laboratory, Department of Public Health, Graduate School of Public Health, Seoul National University, Seoul, Korea
| |
Collapse
|
29
|
Steggink LC, Boer H, Meijer C, Lefrandt JD, Terstappen LWMM, Fehrmann RSN, Gietema JA. Genome-wide association study of cardiovascular disease in testicular cancer patients treated with platinum-based chemotherapy. THE PHARMACOGENOMICS JOURNAL 2020; 21:152-164. [PMID: 33011741 PMCID: PMC7997802 DOI: 10.1038/s41397-020-00191-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 09/04/2020] [Accepted: 09/23/2020] [Indexed: 01/15/2023]
Abstract
Genetic variation may mediate the increased risk of cardiovascular disease (CVD) in chemotherapy-treated testicular cancer (TC) patients compared to the general population. Involved single nucleotide polymorphisms (SNPs) might differ from known CVD-associated SNPs in the general population. We performed an explorative genome-wide association study (GWAS) in TC patients. TC patients treated with platinum-based chemotherapy between 1977 and 2011, age ≤55 years at diagnosis, and ≥3 years relapse-free follow-up were genotyped. Association between SNPs and CVD occurrence during treatment or follow-up was analyzed. Data-driven Expression Prioritized Integration for Complex Trait (DEPICT) provided insight into enriched gene sets, i.e., biological themes. During a median follow-up of 11 years (range 3–37), CVD occurred in 53 (14%) of 375 genotyped patients. Based on 179 SNPs associated at p ≤ 0.001, 141 independent genomic loci associated with CVD occurrence. Subsequent, DEPICT found ten biological themes, with the RAC2/RAC3 network (linked to endothelial activation) as the most prominent theme. Biology of this network was illustrated in a TC cohort (n = 60) by increased circulating endothelial cells during chemotherapy. In conclusion, the ten observed biological themes highlight possible pathways involved in CVD in chemotherapy-treated TC patients. Insight in the genetic susceptibility to CVD in TC patients can aid future intervention strategies.
Collapse
Affiliation(s)
- Lars C Steggink
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Hink Boer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Coby Meijer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Joop D Lefrandt
- Department of Internal Medicine, Division of Vascular Medicine, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Leon W M M Terstappen
- Medical Cell BioPhysics, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
30
|
Trendowski MR, Wheeler HE, El-Charif O, Feldman DR, Hamilton RJ, Vaughn DJ, Fung C, Kollmannsberger C, Einhorn LH, Travis LB, Dolan ME. Clinical and Genome-Wide Analysis of Multiple Severe Cisplatin-Induced Neurotoxicities in Adult-Onset Cancer Survivors. Clin Cancer Res 2020; 26:6550-6558. [PMID: 32998964 DOI: 10.1158/1078-0432.ccr-20-2682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/31/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Cisplatin is a first-line chemotherapeutic for many cancers, but causes neurotoxicity including hearing loss, tinnitus, and peripheral sensory neuropathy. However, no study has comprehensively characterized risk factors for developing multiple (>1) severe neurotoxicities. EXPERIMENTAL DESIGN The relationship between multiple severe neurotoxicities and age, cumulative cisplatin dose, medical history, and lifestyle/behavioral factors was evaluated in 300 cisplatin-treated testicular cancer survivors using logistic regression. Case-control genome-wide association study (GWAS; cases, n = 104 and controls, n = 196) was also performed. RESULTS Age at clinical examination (P = 6.4 × 10-16) and cumulative cisplatin dose (P = 5.4 × 10-4) were positively associated with multiple severe neurotoxicity risk, as were high serum platinum levels (P = 0.02), tobacco use (ever smoker, P = 0.001 and current smoker, P = 0.002), and hypertension (P = 0.01) after adjustment for age and cumulative cisplatin dose. Individuals with multiple severe neurotoxicities were more likely to experience dizziness/vertigo (P = 0.01), Raynaud phenomenon (P = 3.7 × 10-9), and symptoms consistent with peripheral motor neuropathy (P = 4.3 × 10-14) after age and dose adjustment. These patients also reported poorer overall health (P = 2.7 × 10-5) and a greater use of psychotropic medications (P = 0.06). GWAS identified no genome-wide significant SNPs. Gene-based association analysis identified RGS17 (P = 3.9 × 10-5) and FAM20C (P = 5.5 × 10-5) as near genome-wide significant. Decreased FAM20C expression was associated with increased cisplatin sensitivity in tumor cell lines. CONCLUSIONS Certain survivors are more susceptible to cisplatin-induced neurotoxicity, markedly increasing likelihood of developing numerous neuro-otological symptoms that affect quality of life. Genome-wide analysis identified genetic variation in FAM20C as a potentially important risk factor.
Collapse
Affiliation(s)
| | - Heather E Wheeler
- Department of Biology and Program in Bioinformatics, Loyola University Chicago, Chicago, Illinois
| | - Omar El-Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Darren R Feldman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J Hamilton
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David J Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | - Lawrence H Einhorn
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Lois B Travis
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
31
|
Walters CE, Nitin R, Margulis K, Boorom O, Gustavson DE, Bush CT, Davis LK, Below JE, Cox NJ, Camarata SM, Gordon RL. Automated Phenotyping Tool for Identifying Developmental Language Disorder Cases in Health Systems Data (APT-DLD): A New Research Algorithm for Deployment in Large-Scale Electronic Health Record Systems. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2020; 63:3019-3035. [PMID: 32791019 PMCID: PMC7890229 DOI: 10.1044/2020_jslhr-19-00397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/23/2020] [Accepted: 05/19/2020] [Indexed: 05/13/2023]
Abstract
Purpose Data mining algorithms using electronic health records (EHRs) are useful in large-scale population-wide studies to classify etiology and comorbidities (Casey et al., 2016). Here, we apply this approach to developmental language disorder (DLD), a prevalent communication disorder whose risk factors and epidemiology remain largely undiscovered. Method We first created a reliable system for manually identifying DLD in EHRs based on speech-language pathologist (SLP) diagnostic expertise. We then developed and validated an automated algorithmic procedure, called, Automated Phenotyping Tool for identifying DLD cases in health systems data (APT-DLD), that classifies a DLD status for patients within EHRs on the basis of ICD (International Statistical Classification of Diseases and Related Health Problems) codes. APT-DLD was validated in a discovery sample (N = 973) using expert SLP manual phenotype coding as a gold-standard comparison and then applied and further validated in a replication sample of N = 13,652 EHRs. Results In the discovery sample, the APT-DLD algorithm correctly classified 98% (concordance) of DLD cases in concordance with manually coded records in the training set, indicating that APT-DLD successfully mimics a comprehensive chart review. The output of APT-DLD was also validated in relation to independently conducted SLP clinician coding in a subset of records, with a positive predictive value of 95% of cases correctly classified as DLD. We also applied APT-DLD to the replication sample, where it achieved a positive predictive value of 90% in relation to SLP clinician classification of DLD. Conclusions APT-DLD is a reliable, valid, and scalable tool for identifying DLD cohorts in EHRs. This new method has promising public health implications for future large-scale epidemiological investigations of DLD and may inform EHR data mining algorithms for other communication disorders. Supplemental Material https://doi.org/10.23641/asha.12753578.
Collapse
Affiliation(s)
- Courtney E. Walters
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN
- Neuroscience Program, College of Arts and Science, Vanderbilt University, Nashville, TN
| | - Rachana Nitin
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Katherine Margulis
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
- Kennedy Krieger Institute, Baltimore, MD
| | - Olivia Boorom
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Daniel E. Gustavson
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Catherine T. Bush
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Lea K. Davis
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Nancy J. Cox
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Stephen M. Camarata
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Reyna L. Gordon
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Fung C, Dinh PC, Fossa SD, Travis LB. Testicular Cancer Survivorship. J Natl Compr Canc Netw 2020; 17:1557-1568. [PMID: 31805527 DOI: 10.6004/jnccn.2019.7369] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/14/2019] [Indexed: 11/17/2022]
Abstract
Testicular cancer (TC) is the most common cancer among men aged 18 to 39 years. It is highly curable, with a 10-year relative survival approaching 95% due to effective cisplatin-based chemotherapy. Given the increasing incidence of TC and improved survival, TC survivors (TCS) now account for approximately 4% of all US male cancer survivors. They have also become a valuable cohort for adult-onset cancer survivorship research, given their prolonged survival. Commensurately, long-term treatment-related complications have emerged as important survivorship issues. These late effects include life-threatening conditions, such as second malignant neoplasms and cardiovascular disease. Moreover, TCS can also experience hearing loss, tinnitus, neurotoxicity, nephrotoxicity, pulmonary toxicity, hypogonadism, infertility, anxiety, depression, cognitive impairment, and chronic cancer-related fatigue. Characterization of the number and severity of long-term adverse health outcomes among TCS remains critical to develop risk-stratified, evidence-based follow-up guidelines and to inform the development of preventive measures and interventions. In addition, an improved understanding of the long-term effects of TC treatment on mortality due to noncancer causes and second malignant neoplasms remains paramount. Future research should focus on the continued development of large, well-characterized clinical cohorts of TCS for lifelong follow-up. These systematic, comprehensive approaches can provide the needed infrastructure for further investigation of long-term latency patterns of various medical and psychosocial morbidities and for more in-depth studies investigating associated etiopathogenetic pathways. Studies examining premature physiologic aging may also serve as new frontiers in TC survivorship research.
Collapse
Affiliation(s)
- Chunkit Fung
- aUniversity of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Paul C Dinh
- bIndiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana.,cDepartment of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana; and
| | | | - Lois B Travis
- bIndiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| |
Collapse
|
35
|
Peng H, Wu X, Wen Y, Li C, Lin J, Li J, Xiong S, Zhong R, Liang H, Cheng B, Liu J, He J, Liang W. Association between systemic sclerosis and risk of lung cancer: results from a pool of cohort studies and Mendelian randomization analysis. Autoimmun Rev 2020; 19:102633. [PMID: 32801043 DOI: 10.1016/j.autrev.2020.102633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Population-based cohort studies have indicated that systemic sclerosis (SSc) may be associated with an increased risk of lung cancer. However, there are few studies that comprehensively investigate their correlation and the causal effect remains unknown. METHODS A systematic search of PubMed, Web of Science, Cochrane Library and Embase from the inception dates to December 1, 2019 was carried out. Meta-analysis was performed to calculate odds ratio (OR) and corresponding 95% confidence interval (CI) using random-effects models. Subgroup analyses were performed regarding gender. Two-sample Mendelian randomization (MR) was carried out with summary data from published genome-wide association studies of SSc (Neale Lab, 3871 individuals; UK Biobank, 463,315 individuals) and lung cancer (International Lung Cancer Consortium, 27,209 individuals; UK Biobank, 508,977 individuals). Study-specific estimates were summarized using inverse variance-weighted, weighted median, and MR-Egger method. RESULTS Through meta-analysis of 10 population-based cohort studies involving 12,218 patients, we observed a significantly increased risk of lung cancer among patients with SSc (OR 2.80, 95% CI 1.55-5.03). In accordance with subgroup analysis, male patients (OR 4.11, 95% CI 1.92-8.79) had a 1.5-fold higher lung cancer risk compared with female patients (OR 2.73, 95% CI 1.41-5.27). However, using a score of 11 SSc-related single nucleotide polymorphisms (p < 5*10-8) as instrumental variables, the MR study did not support a causality between SSc and lung cancer (OR 1.001, 95% CI 0.929-1.100, p = 0.800). Specifically, subgroup MR analyses indicated that SSc was not associated with increased risks of non-small-cell lung cancer (OR 1.000, 95% CI 0.999-1.000, p = 0.974), including lung adenocarcinoma (OR 0.996, 95% CI 0.906-1.094, p = 0.927), squamous cell lung carcinoma (OR 1.034, 95% CI 0.937-1.140, p = 0.507), nor small-cell lung cancer (OR 1.000, 95% CI 0.999-1.000, p = 0.837). CONCLUSIONS This study indicated an increased risk of lung cancer among patients with SSc by meta-analysis, whereas the MR study did not support a causality between the two diseases. Further studies are warranted to investigate the factors underlying the attribution of SSc to lung cancer risk.
Collapse
Affiliation(s)
- Haoxin Peng
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou 511436, China
| | - Xiangrong Wu
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou 511436, China
| | - Yaokai Wen
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou 511436, China
| | - Caichen Li
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinsheng Lin
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou 511436, China
| | - Jianfu Li
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shan Xiong
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ran Zhong
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hengrui Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bo Cheng
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun Liu
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
36
|
Bernsen EC, Hagleitner MM, Kouwenberg TW, Hanff LM. Pharmacogenomics as a Tool to Limit Acute and Long-Term Adverse Effects of Chemotherapeutics: An Update in Pediatric Oncology. Front Pharmacol 2020; 11:1184. [PMID: 32848787 PMCID: PMC7421781 DOI: 10.3389/fphar.2020.01184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
In the past decades, new cancer treatments have been introduced in pediatric oncology leading to improvement in clinical outcomes and survival rates. However, due to inter-individual differences, some children experience severe chemotherapy-induced toxicities or a poor clinical outcome. An explanation for the diversity in response to chemotherapy is genetic variation, leading to differences in expression and activity of metabolizing and transport enzymes as well as drug targets. Pharmacogenetic testing has emerged as a promising tool to predict and limit acute and long-term adverse effects in patients. However, in pediatric oncology, limited number of patients and a considerable diversity in study results complicate the interpretation of test results and its clinical relevance. With this review, we provide an overview of new developments over the past four years regarding relevant polymorphisms related to toxicity in pediatric oncology. The following chemotherapeutics and associated toxicities are discussed: alkylating agents, anthracyclines, asparaginase, methotrexate, platinum compounds, steroids, thiopurines, topoisomerase inhibitors, and vinca alkaloids. Our review identifies several questions regarding the role of genetic variants in chemotherapy-induced toxicities. Ambiguities in the literature stem from small population sizes, differences in (statistical) interpretation and variations in sequencing technologies as well as different clinical outcome definitions. Standardization of clinical outcome data and toxicity definitions within electronic health records combined with the increased availability of genomic sequence techniques in clinical practice will help to validate these models in upcoming years.
Collapse
Affiliation(s)
- Emma C. Bernsen
- Pharmacy, Princess Máxima Centre for Pediatric Oncology, Utrecht, Netherlands
| | - Melanie M. Hagleitner
- Department of Pediatric Hemato-oncology, Princess Máxima Centre for Pediatric Oncology, Utrecht, Netherlands
| | - Theodorus W. Kouwenberg
- Department of Pediatric Hemato-oncology, Princess Máxima Centre for Pediatric Oncology, Utrecht, Netherlands
| | - Lidwien M. Hanff
- Pharmacy, Princess Máxima Centre for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
37
|
Ben Ayed W, Ben Said A, Hamdi A, Mokrani A, Masmoudi Y, Toukabri I, Limayem I, Yahyaoui Y. Toxicity, risk factors and management of cisplatin-induced toxicity: A prospective study. J Oncol Pharm Pract 2020; 26:1621-1629. [DOI: 10.1177/1078155219901305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background and propose Cisplatin is a cytotoxic drug that triggers several toxicities. However, nephrotoxicity and ototoxicity remain major clinical limitations. The aim of our study was to evaluate the incidence of chemotherapy toxicity induced by cisplatin and to analyze the influence of risk factors in the Tunisian population. Methods We performed a prospective descriptive study in a period of four months. Patients were eligible if they had pathologically confirmed malignancies and treated with cisplatin-regimen chemotherapy. Nephrotoxicity and digestive toxicity were graded according to the World Health Organization toxicity scale and ototoxicity was scored clinically according to the Common Terminology Criteria for Adverse Events (CTCAE). Multivariate logistic regression analysis was performed to evaluate the influence of clinical variables on cisplatin-induced toxicity. Results A total of 150 patients were included. Forty-four percent of patients developed cisplatin-regimen toxicity: 15% developed cisplatin-induced nephrotoxicity, 9% cisplatin-induced ototoxicity and 27% digestive toxicity. In the multivariate analysis, age >65 years (OR= 6.129, p = 0.010), metastatic cancer (OR = 0.171, p = 0.007) and cumulative dose (OR= 1.004 mg/m2; p = 0.042) were strong predisposing factors for CDDP-induced nephrotoxicity. The cumulative dose was an independent prognostic indicator for digestive toxicity (OR = 0.997, p = 0.002). Conclusion In our study, age >65 years and metastatic cancer were risk factors for cisplatin-induced nephrotoxicities. We also found the correlation between cumulative dose and nephrotoxicity or digestive toxicity.
Collapse
Affiliation(s)
- Wiem Ben Ayed
- Department of Pharmacy, Salah Azaiez Institute, Tunis, Tunisia
| | - Azza Ben Said
- Department of Pharmacy, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Adel Hamdi
- Department of Pharmacy, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Amina Mokrani
- Department of Oncology, Salah Azaiez Institute, Tunis, Tunisia
| | - Yosri Masmoudi
- Department of Pharmacy, Salah Azaiez Institute, Tunis, Tunisia
| | - Imen Toukabri
- Department of Pharmacy, Salah Azaiez Institute, Tunis, Tunisia
| | - Imen Limayem
- Department of Pharmacy, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Yosra Yahyaoui
- Department of Oncology, Salah Azaiez Institute, Tunis, Tunisia
| |
Collapse
|
38
|
Baguley DM, Prayuenyong P. Looking beyond the audiogram in ototoxicity associated with platinum-based chemotherapy. Cancer Chemother Pharmacol 2019; 85:245-250. [PMID: 31865419 PMCID: PMC7015967 DOI: 10.1007/s00280-019-04012-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022]
Abstract
Introduction Ototoxicity associated with platinum-based chemotherapy is highly prevalent and can cause detrimental consequences among cancer survivors. Discussion In this article, we highlight important aspects of the evaluation of ototoxicity with the aim to increase awareness of Oncologists in this regard. Standard pure tone audiometry alone is inadequate for this context. Comprehensive and consistent hearing tests should be implemented in a monitoring and surveillance program. High-frequency audiometry (10–16 kHz) is a sensitive tool in the detection of ototoxic hearing loss at onset. In addition to threshold audiometry, measures of speech comprehension (both in quiet and in noise) can add useful information in the evaluation of hearing in real-life situations. Not only hearing loss, but also tinnitus and imbalance are common in patients who receive platinum-based chemotherapy, and can cause debilitating effects upon quality of life in this population. Moreover, self-report measures associated with cochlear and vestibular handicaps can provide valuable information regarding the impact of ototoxicity. Conclusions It is vital to build awareness about the variety and impact of the symptoms of ototoxicity. Comprehensive evaluation of hearing status along with self-reported impact of the cochlear and vestibular handicap should be implemented in a monitoring and surveillance program for appropriate investigation and management.
Collapse
Affiliation(s)
- David M Baguley
- Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Ropewalk House, 113 The Ropewalk, Nottingham, UK.,Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Pattarawadee Prayuenyong
- Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK. .,NIHR Nottingham Biomedical Research Centre, Ropewalk House, 113 The Ropewalk, Nottingham, UK. .,Nottingham University Hospitals NHS Trust, Nottingham, UK. .,Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand.
| |
Collapse
|
39
|
Turan C, Kantar M, Aktan Ç, Kosova B, Orman M, Bilgen C, Kirazlı T. Cisplatin ototoxicity in children: risk factors and its relationship with polymorphisms of DNA repair genes ERCC1, ERCC2, and XRCC1. Cancer Chemother Pharmacol 2019; 84:1333-1338. [PMID: 31586226 DOI: 10.1007/s00280-019-03968-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/25/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE We aimed to investigate the cisplatin-related hearing toxicity and its possible relationship with polymorphic variants in DNA repair genes, ERCC1, ERCC2, and XRCC1. METHODS Fifty patients treated with cisplatin in the past were included in the study. There were 29 females and 21 males; mean age 13.4 ± 6.0 years). The polymorphism in DNA repair genes was studied using primer and probes in Light Cycler device after DNA isolation was carried out with PCR technique. The polymorphisms and clinical risk factors were evaluated using Chi square test and logistic regression modelling. RESULTS The patients had hearing loss in 44%. For ERCC1 gene, the patients with hearing loss had 50% of GG (wild type), 40.9% of AG and 9.1% of AA genotypes, while the patients without hearing loss had 28.6% of GG, 53.5% of AG, and 17.9% of AA genotypes. For ERCC2 gene, the patients with hearing loss had 18.2% of GG (wild type), 40.9% of TG, and 40.9% of TT genotypes, while the patients without hearing loss had 10.7% of GG 39.3% of TG, and 50% of TT genotypes. For XRCC1 gene, the patients with hearing loss had 18.2% of CC (wild type), 59.1% of CT, and 22.7% of TT genotypes, while the patients without hearing loss had 35.7% of CC, 50% of CT, and 14.3% of TT genotypes. There was no statistically significant association among the groups (p = 0.24). CONCLUSION We did not find a relationship between DNA repair gene polymorphisms and hearing toxicity of cisplatin.
Collapse
Affiliation(s)
- Caner Turan
- Department of Pediatrics, Ege University School of Medicine, Izmir, Turkey
| | - Mehmet Kantar
- Department of Pediatrics, Division of Pediatric Oncology, Ege University School of Medicine, Izmir, Turkey.
| | - Çağdaş Aktan
- Department of Medical Biology, Beykent University School of Medicine, Istanbul, Turkey
| | - Buket Kosova
- Department of Medical Biology, Beykent University School of Medicine, Istanbul, Turkey
| | - Mehmet Orman
- Department of Biostatistics and Medical Informatics, Ege University School of Medicine, Izmir, Turkey
| | - Cem Bilgen
- Department of Otorhinolaryngology, Ege University School of Medicine, Izmir, Turkey
| | - Tayfun Kirazlı
- Department of Otorhinolaryngology, Ege University School of Medicine, Izmir, Turkey
| |
Collapse
|
40
|
Trendowski MR, El-Charif O, Ratain MJ, Monahan P, Mu Z, Wheeler HE, Dinh PC, Feldman DR, Ardeshir-Rouhani-Fard S, Hamilton RJ, Vaughn DJ, Fung C, Kollmannsberger C, Mushiroda T, Kubo M, Hannigan R, Strathmann F, Einhorn LH, Fossa SD, Travis LB, Dolan ME. Clinical and Genome-Wide Analysis of Serum Platinum Levels after Cisplatin-Based Chemotherapy. Clin Cancer Res 2019; 25:5913-5924. [PMID: 31296530 PMCID: PMC6774840 DOI: 10.1158/1078-0432.ccr-19-0113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/17/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Serum platinum is measurable for years after completion of cisplatin-based chemotherapy (CBC). We report the largest investigation of serum platinum levels to date of 1,010 testicular cancer survivors (TCS) assessed 1-35 years after CBC and evaluate genetic contributions to these levels. EXPERIMENTAL DESIGN Eligible TCS given 300 or 400 (±15) mg/m2 cisplatin underwent extensive audiometric testing, clinical examination, completed questionnaires, and had crude serum platinum levels measured. Associations between serum platinum and various risk factors and toxicities were assessed after fitting a biexponential model adjusted for follow-up time and cumulative cisplatin dose. A genome-wide association study (GWAS) was performed using the serum platinum residuals of the dose and time-adjusted model. RESULTS Serum platinum levels exceeded the reference range for approximately 31 years, with a strong inverse relationship with creatinine clearance at follow-up (age-adjusted P = 2.13 × 10-3). We observed a significant, positive association between residual platinum values and luteinizing hormone (age-adjusted P = 6.58 × 10-3). Patients with high residual platinum levels experienced greater Raynaud phenomenon than those with medium or low levels (age-adjusted ORhigh/low = 1.46; P = 0.04), as well as a higher likelihood of developing tinnitus (age-adjusted ORhigh/low = 1.68, P = 0.07). GWAS identified one single-nucleotide polymorphism (SNP) meeting genome-wide significance, rs1377817 (P = 4.6 × 10-8, a SNP intronic to MYH14). CONCLUSIONS This study indicates that residual platinum values are correlated with several cisplatin-related toxicities. One genetic variant is associated with these levels.
Collapse
Affiliation(s)
| | - Omar El-Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Patrick Monahan
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Zepeng Mu
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Heather E Wheeler
- Departments of Biology and Computer Science, Loyola University Chicago, Chicago, Illinois
| | - Paul C Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Darren R Feldman
- Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - Robert J Hamilton
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David J Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - Robyn Hannigan
- School for the Environment, University of Massachusetts Boston, Boston, Massachusetts
| | | | - Lawrence H Einhorn
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Sophie D Fossa
- Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway
| | - Lois B Travis
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana.
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
41
|
Zaid MA, Gathirua-Mwangi WG, Fung C, Monahan PO, El-Charif O, Williams AM, Feldman DR, Hamilton RJ, Vaughn DJ, Beard CJ, Cook R, Althouse SK, Ardeshir-Rouhani-Fard S, Dinh PC, Sesso HD, Einhorn LH, Fossa SD, Travis LB. Clinical and Genetic Risk Factors for Adverse Metabolic Outcomes in North American Testicular Cancer Survivors. J Natl Compr Canc Netw 2019. [PMID: 29523664 DOI: 10.6004/jnccn.2017.7046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Testicular cancer survivors (TCS) are at significantly increased risk for cardiovascular disease (CVD), with metabolic syndrome (MetS) an established risk factor. No study has addressed clinical and genetic MetS risk factors in North American TCS. Patients and Methods: TCS were aged <55 years at diagnosis and received first-line chemotherapy. Patients underwent physical examination, and had lipid panels, testosterone, and soluble cell adhesion molecule-1 (sICAM-1) evaluated. A single nucleotide polymorphism in rs523349 (5-α-reductase gene, SRD5A2), recently implicated in MetS risk, was genotyped. Using standard criteria, MetS was defined as ≥3 of the following: hypertension, abdominal obesity, hypertriglyceridemia, decreased high-density lipoprotein (HDL) cholesterol level, and diabetes. Matched controls were derived from the National Health and Nutrition Examination Survey. Results: We evaluated 486 TCS (median age, 38.1 years). TCS had a higher prevalence of hypertension versus controls (43.2% vs 30.7%; P<.001) but were less likely to have decreased HDL levels (23.7% vs 34.8%; P<.001) or abdominal obesity (28.2% vs 40.1%; P<.001). Overall MetS frequency was similar in TCS and controls (21.0% vs 22.4%; P=.59), did not differ by treatment (P=.20), and was not related to rs523349 (P=.61). For other CVD risk factors, TCS were significantly more likely to have elevated low-density lipoprotein (LDL) cholesterol levels (17.7% vs 9.3%; P<.001), total cholesterol levels (26.3% vs 11.1%; P<.001), and body mass index ≥25 kg/m2 (75.1% vs 69.1%; P=.04). On multivariate analysis, age at evaluation (P<.001), testosterone level ≤3.0 ng/mL (odds ratio [OR], 2.06; P=.005), and elevated sICAM-1 level (ORhighest vs lowest quartile, 3.58; P=.001) were significantly associated with MetS. Conclusions and Recommendations: Metabolic abnormalities in TCS are characterized by hypertension and increased LDL and total cholesterol levels but lower rates of decreased HDL levels and abdominal obesity, signifying possible shifts in fat distribution and fat metabolism. These changes are accompanied by hypogonadism and inflammation. TCS have a high prevalence of CVD risk factors that may not be entirely captured by standard MetS criteria. Cancer treatment-associated MetS requires further characterization.
Collapse
|
42
|
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
|
43
|
Abu Zaid M, Dinh PC, Monahan PO, Fung C, El-Charif O, Feldman DR, Hamilton RJ, Vaughn DJ, Beard CJ, Cook R, Althouse S, Ardeshir-Rouhani-Fard S, Sesso HD, Huddart R, Mushiroda T, Kubo M, Eileen Dolan M, Einhorn LH, Fossa SD, Travis LB. Adverse Health Outcomes in Relationship to Hypogonadism After Chemotherapy: A Multicenter Study of Testicular Cancer Survivors. J Natl Compr Canc Netw 2019; 17:459-468. [PMID: 31085753 PMCID: PMC6712564 DOI: 10.6004/jnccn.2018.7109] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND This study examined the prevalence of hypogonadism, its clinical and genetic risk factors, and its relationship to adverse health outcomes (AHOs) in North American testicular cancer survivors (TCS) after modern platinum-based chemotherapy. PATIENTS AND METHODS Eligible TCS were <55 years of age at diagnosis and treated with first-line platinum-based chemotherapy. Participants underwent physical examinations and completed questionnaires regarding 15 AHOs and health behaviors. Hypogonadism was defined as serum testosterone levels ≤3.0 ng/mL or use of testosterone replacement therapy. We investigated the role of 2 single nucleotide polymorphisms (rs6258 and rs12150660) in the sex hormone-binding globulin (SHBG) locus implicated in increased hypogonadism risk in the general population. RESULTS Of 491 TCS (median age at assessment, 38.2 years; range, 18.7-68.4 years), 38.5% had hypogonadism. Multivariable binary logistic regression analysis identified hypogonadism risk factors, including age at clinical evaluation (odds ratio [OR], 1.42 per 10-year increase; P= .006) and body mass index of 25 to <30 kg/m2 (OR, 2.08; P= .011) or ≥30 kg/m2 (OR, 2.36; P= .005) compared with <25 kg/m2. TCS with ≥2 risk alleles for the SHBG SNPs had a marginally significant increased hypogonadism risk (OR, 1.45; P= .09). Vigorous-intensity physical activity appeared protective (OR, 0.66; P= .07). Type of cisplatin-based chemotherapy regimen and socioeconomic factors did not correlate with hypogonadism. Compared with TCS without hypogonadism, those with hypogonadism were more likely to report ≥2 AHOs (65% vs 51%; P= .003), to take medications for hypercholesterolemia (20.1% vs 6.0%; P<.001) or hypertension (18.5% vs 10.6%; P= .013), and to report erectile dysfunction (19.6% vs 11.9%; P= .018) or peripheral neuropathy (30.7% vs 22.5%; P= .041). A marginally significant trend for increased use of prescription medications for either diabetes (5.8% vs 2.6%; P= .07) or anxiety/depression (14.8% vs 9.3%; P= .06) was observed. CONCLUSIONS At a relatively young median age, more than one-third of TCS have hypogonadism, which is significantly associated with increased cardiovascular disease risk factors, and erectile dysfunction. Providers should screen TCS for hypogonadism and treat symptomatic patients.
Collapse
Affiliation(s)
- Mohammad Abu Zaid
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Paul C. Dinh
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Patrick O. Monahan
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Chunkit Fung
- University of Rochester Medical Center, James P. Wilmot Cancer Institute, Rochester, New York
| | - Omar El-Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Darren R. Feldman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NewYork
| | | | - David J. Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Clair J. Beard
- Department of Radiation Oncology, Dana-FarberCancer Institute
| | - Ryan Cook
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Sandra Althouse
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | | | - Howard D. Sesso
- Divisions of Preventive Medicine andAging, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Taisei Mushiroda
- The RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | | | - M. Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Lawrence H. Einhorn
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Sophie D. Fossa
- Department of Oncology, Oslo University Hospital, Radium Hospital, Oslo, Norway
| | - Lois B. Travis
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | | |
Collapse
|
44
|
Driessen CM, Ham JC, Te Loo M, van Meerten E, van Lamoen M, Hakobjan MH, Takes RP, van der Graaf WT, Kaanders JH, Coenen MJH, van Herpen CM. Genetic Variants as Predictive Markers for Ototoxicity and Nephrotoxicity in Patients with Locally Advanced Head and Neck Cancer Treated with Cisplatin-Containing Chemoradiotherapy (The PRONE Study). Cancers (Basel) 2019; 11:cancers11040551. [PMID: 30999660 PMCID: PMC6520709 DOI: 10.3390/cancers11040551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/20/2019] [Accepted: 04/15/2019] [Indexed: 01/11/2023] Open
Abstract
Ototoxicity and nephrotoxicity are potentially irreversible side effects of chemoradiotherapy with cisplatin in locally advanced head and neck cancer (LAHNC) patients. Several predictive genetic variants have been described, but as yet none in LAHNC patients. The aim of this study is to investigate genetic variants as predictors for ototoxicity and nephrotoxicity in LAHNC patients treated with cisplatin-containing chemoradiotherapy. Our prospective cohort of 92 patients was genotyped for 10 genetic variants and evaluated for their association with cisplatin-induced ototoxicity (ACYP2, COMT, TPMT and WFS1) and nephrotoxicity (OCT2, MATE and XPD). Ototoxicity was determined by patient-reported complaints as well as tone audiometrical assessments. Nephrotoxicity was defined as a decrease of ≥25% in creatinine clearance during treatment compared to baseline. A significant association was observed between carriership of the A allele for rs1872328 in the ACYP2 gene and cisplatin-induced clinically determined ototoxicity (p = 0.019), and not for ototoxicity measured by tone audiometrical assessments (p = 0.449). Carriership of a T allele for rs316019 in the OCT2 gene was significantly associated with nephrotoxicity at any time during chemoradiotherapy (p = 0.022), but not with nephrotoxicity at the end of the chemoradiotherapy. In conclusion, we showed prospectively that in LAHNC patients genetic variants in ACYP2 are significantly associated with clinically determined ototoxicity. Validation studies are necessary to prove the added value for individualized treatments plans in these patients.
Collapse
Affiliation(s)
- Chantal M Driessen
- Department of Medical Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Janneke C Ham
- Department of Medical Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Maroeska Te Loo
- Department of Pediatric Hematology and Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Postbox 2040, 3000 CA Rotterdam, The Netherlands.
| | - Maurits van Lamoen
- Department of Otorhinolaryngology and Head and Neck Surgery, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Marina H Hakobjan
- Department of Human Genetics, Radboud Institute of Health Sciences, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Robert P Takes
- Department of Otorhinolaryngology and Head and Neck Surgery, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Winette T van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Johannes H Kaanders
- Department of Radiation Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Marieke J H Coenen
- Department of Human Genetics, Radboud Institute of Health Sciences, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Carla M van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
45
|
El Charif O, Mapes B, Trendowski MR, Wheeler HE, Wing C, Dinh PC, Frisina RD, Feldman DR, Hamilton RJ, Vaughn DJ, Fung C, Kollmannsberger C, Mushiroda T, Kubo M, Gamazon ER, Cox NJ, Huddart R, Ardeshir-Rouhani-Fard S, Monahan P, Fossa SD, Einhorn LH, Travis LB, Dolan ME. Clinical and Genome-wide Analysis of Cisplatin-induced Tinnitus Implicates Novel Ototoxic Mechanisms. Clin Cancer Res 2019; 25:4104-4116. [PMID: 30952644 DOI: 10.1158/1078-0432.ccr-18-3179] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/13/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Cisplatin, a commonly used chemotherapeutic, results in tinnitus, the phantom perception of sound. Our purpose was to identify the clinical and genetic determinants of tinnitus among testicular cancer survivors (TCS) following cisplatin-based chemotherapy. EXPERIMENTAL DESIGN TCS (n = 762) were dichotomized to cases (moderate/severe tinnitus; n = 154) and controls (none; n = 608). Logistic regression was used to evaluate associations with comorbidities and SNP dosages in genome-wide association study (GWAS) following quality control and imputation (covariates: age, noise exposure, cisplatin dose, genetic principal components). Pathway over-representation tests and functional studies in mouse auditory cells were performed. RESULTS Cisplatin-induced tinnitus (CisIT) significantly associated with age at diagnosis (P = 0.007) and cumulative cisplatin dose (P = 0.007). CisIT prevalence was not significantly greater in 400 mg/m2-treated TCS compared with 300 (P = 0.41), but doses >400 mg/m2 (median 580, range 402-828) increased risk by 2.61-fold (P < 0.0001). CisIT cases had worse hearing at each frequency (0.25-12 kHz, P < 0.0001), and reported more vertigo (OR = 6.47; P < 0.0001) and problems hearing in a crowd (OR = 8.22; P < 0.0001) than controls. Cases reported poorer health (P < 0.0001) and greater psychotropic medication use (OR = 2.4; P = 0.003). GWAS suggested a variant near OTOS (rs7606353, P = 2 × 10-6) and OTOS eQTLs were significantly enriched independently of that SNP (P = 0.018). OTOS overexpression in HEI-OC1, a mouse auditory cell line, resulted in resistance to cisplatin-induced cytotoxicity. Pathway analysis implicated potassium ion transport (q = 0.007). CONCLUSIONS CisIT associated with several neuro-otological symptoms, increased use of psychotropic medication, and poorer health. OTOS, expressed in the cochlear lateral wall, was implicated as protective. Future studies should investigate otoprotective targets in supporting cochlear cells.
Collapse
Affiliation(s)
- Omar El Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Brandon Mapes
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Heather E Wheeler
- Departments of Biology and Computer Science, Loyola University Chicago, Chicago, Illinois
| | - Claudia Wing
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Paul C Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Robert D Frisina
- Departments of Medical and Chemical & Biomolecular Engineering and Communication Sciences & Disorders, Global Center for Hearing and Speech Research, University of South Florida, Tampa, Florida
| | - Darren R Feldman
- Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Robert J Hamilton
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David J Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - Eric R Gamazon
- Clare Hall, University of Cambridge, Cambridge, United Kingdom.,Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nancy J Cox
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Patrick Monahan
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Sophie D Fossa
- Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway
| | - Lawrence H Einhorn
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Lois B Travis
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana.
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
46
|
Tserga E, Nandwani T, Edvall NK, Bulla J, Patel P, Canlon B, Cederroth CR, Baguley DM. The genetic vulnerability to cisplatin ototoxicity: a systematic review. Sci Rep 2019; 9:3455. [PMID: 30837596 PMCID: PMC6401165 DOI: 10.1038/s41598-019-40138-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Ototoxicity is one of the major side-effects of platinum-based chemotherapy, in particular cisplatin (cis-diammine dichloroplatinum II). To our knowledge, no systematic review has previously provided a quantitative summary estimate of the impact of genetics upon the risk of developing hearing loss. We searched Embase, Medline, ASSIA, Pubmed, Scopus, and Web of Science, for studies documenting the genetic risk of ototoxicity in patients with cancer treated with cisplatin. Titles/abstracts and full texts were reviewed for inclusion. Meta-analytic estimates of risk (Odds Ratio) from the pooled data were calculated for studies that have been repeated twice or more. The search identified 3891 papers, of which 30 were included. The majority were retrospective (44%), ranging from n = 39 to n = 317, some including only patients younger than 25 years of age (33%), and some on both genders (80%). The most common cancers involved were osteosarcoma (53%), neuroblastoma (37%), prostate (17%) and reproductive (10%). Most studies performed genotyping, though only 5 studies performed genome-wide association studies. Nineteen single-nucleotide polymorphisms (SNPs) from 15 genes were repeated more than twice. Meta-analysis of group data indicated that rs1872328 on ACYP2, which plays a role in calcium homeostasis, increases the risk of ototoxicity by 4.61 (95% CI: 3.04-7.02; N = 696, p < 0.0001) as well as LRP2 rs4668123 shows a cumulated Odds Ratio of 3.53 (95% CI: 1.48-8.45; N = 118, p = 0.0059), which could not be evidenced in individual studies. Despite the evidence of heterogeneity across studies, these meta-analytic results from 30 studies are consistent with a view of a genetic predisposition to platinum-based chemotherapy mediated ototoxicity. These new findings are informative and encourage the genetic screening of cancer patients in order to identify patients with greater vulnerability of developing hearing loss, a condition having a potentially large impact on quality of life. More studies are needed, with larger sample size, in order to identify additional markers of ototoxic risk associated with platinum-based chemotherapy and investigate polygenic risks, where multiple markers may exacerbate the side-effects.
Collapse
Affiliation(s)
- Evangelia Tserga
- Experimental Audiology, Biomedicum, Karolinska Institutet, Solnavägen 9, 171 65, Stockholm, Sweden
| | - Tara Nandwani
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Niklas K Edvall
- Experimental Audiology, Biomedicum, Karolinska Institutet, Solnavägen 9, 171 65, Stockholm, Sweden
| | - Jan Bulla
- Department of Mathematics, University of Bergen, Bergen, Norway.,Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstraße 84, 93053, Regensburg, Germany
| | - Poulam Patel
- Division of Oncology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Barbara Canlon
- Experimental Audiology, Biomedicum, Karolinska Institutet, Solnavägen 9, 171 65, Stockholm, Sweden
| | - Christopher R Cederroth
- Experimental Audiology, Biomedicum, Karolinska Institutet, Solnavägen 9, 171 65, Stockholm, Sweden
| | - David M Baguley
- Otology and Hearing Group, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK. .,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| |
Collapse
|
47
|
Morton LM, Kerns SL, Dolan ME. Role of Germline Genetics in Identifying Survivors at Risk for Adverse Effects of Cancer Treatment. Am Soc Clin Oncol Educ Book 2018; 38:775-786. [PMID: 30231410 DOI: 10.1200/edbk_201391] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The growing population of cancer survivors often faces adverse effects of treatment, which have a substantial impact on morbidity and mortality. Although certain adverse effects are thought to have a significant heritable component, much work remains to be done to understand the role of germline genetic factors in the development of treatment-related toxicities. In this article, we review current understanding of genetic susceptibility to a range of adverse outcomes among cancer survivors (e.g., fibrosis, urinary and rectal toxicities, ototoxicity, chemotherapy-induced peripheral neuropathy, subsequent malignancies). Most previous research has been narrowly focused, investigating variation in candidate genes and pathways such as drug metabolism, DNA damage and repair, and inflammation. Few of the findings from these earlier candidate gene studies have been replicated in independent populations. Advances in understanding of the genome, improvements in technology, and reduction in laboratory costs have led to recent genome-wide studies, which agnostically interrogate common and/or rare variants across the entire genome. Larger cohorts of patients with homogeneous treatment exposures and systematic ascertainment of well-defined outcomes as well as replication in independent study populations are essential aspects of the study design and are increasingly leading to the discovery of variants associated with each of the adverse outcomes considered in this review. In the long-term, validated germline genetic associations hold tremendous promise for more precisely identifying patients at highest risk for developing adverse treatment effects, with implications for frontline therapy decision-making, personalization of long-term follow-up guidelines, and potential identification of targets for prevention or treatment of the toxicity.
Collapse
Affiliation(s)
- Lindsay M Morton
- From the Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute at the National Institutes of Health, Bethesda, MD; Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY; Department of Medicine, University of Chicago, Chicago, IL
| | - Sarah L Kerns
- From the Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute at the National Institutes of Health, Bethesda, MD; Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY; Department of Medicine, University of Chicago, Chicago, IL
| | - M Eileen Dolan
- From the Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute at the National Institutes of Health, Bethesda, MD; Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY; Department of Medicine, University of Chicago, Chicago, IL
| |
Collapse
|
48
|
Trendowski MR, El Charif O, Dinh PC, Travis LB, Dolan ME. Genetic and Modifiable Risk Factors Contributing to Cisplatin-induced Toxicities. Clin Cancer Res 2018; 25:1147-1155. [PMID: 30305294 DOI: 10.1158/1078-0432.ccr-18-2244] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 09/07/2018] [Accepted: 10/05/2018] [Indexed: 12/22/2022]
Abstract
Effective administration of traditional cytotoxic chemotherapy is often limited by off-target toxicities. This clinical dilemma is epitomized by cisplatin, a platinating agent, which has potent antineoplastic activity due to its affinity for DNA and other intracellular nucleophiles. Despite its efficacy against many adult-onset and pediatric malignancies, cisplatin elicits multiple off-target toxicities that can not only severely impact a patient's quality of life but also lead to dose reductions or the selection of alternative therapies that can ultimately affect outcomes. Without an effective therapeutic measure by which to successfully mitigate many of these symptoms, there have been attempts to identify a priori those individuals who are more susceptible to developing these sequelae through studies of genetic and nongenetic risk factors. Older age is associated with cisplatin-induced ototoxicity, neurotoxicity, and nephrotoxicity. Traditional genome-wide association studies have identified single-nucleotide polymorphisms in ACYP2 and WFS1 associated with cisplatin-induced hearing loss. However, validating associations between specific genotypes and cisplatin-induced toxicities with enough stringency to warrant clinical application remains challenging. This review summarizes the current state of knowledge with regard to specific adverse sequelae following cisplatin-based therapy, with a focus on ototoxicity, neurotoxicity, nephrotoxicity, myelosuppression, and nausea/emesis. We discuss variables (genetic and nongenetic) contributing to these detrimental toxicities and currently available means to prevent or treat their occurrence.
Collapse
Affiliation(s)
- Matthew R Trendowski
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Omar El Charif
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Paul C Dinh
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Lois B Travis
- Indiana University, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - M Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
49
|
Abstract
Testicular cancer is the most common malignancy among men between 14 and 44 years of age, and its incidence has risen over the past two decades in Western countries. Both genetic and environmental factors contribute to the development of testicular cancer, for which cryptorchidism is the most common risk factor. Progress has been made in our understanding of the disease since the initial description of carcinoma in situ of the testis in 1972 (now referred to as germ cell neoplasia in situ), which has led to improved treatment options. The combination of surgery and cisplatin-based chemotherapy has resulted in a cure rate of >90% in patients with testicular cancer, although some patients become refractory to chemotherapy or have a late relapse; an improved understanding of the molecular determinants underlying tumour sensitivity and resistance may lead to the development of novel therapies for these patients. This Primer provides an overview of the biology, epidemiology, diagnosis and current treatment guidelines for testicular cancer, with a focus on germ cell tumours. We also outline areas for future research and what to expect in the next decade for testicular cancer.
Collapse
|
50
|
Phenotype-Specific Enrichment of Mendelian Disorder Genes near GWAS Regions across 62 Complex Traits. Am J Hum Genet 2018; 103:535-552. [PMID: 30290150 DOI: 10.1016/j.ajhg.2018.08.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/28/2018] [Indexed: 01/29/2023] Open
Abstract
Although recent studies provide evidence for a common genetic basis between complex traits and Mendelian disorders, a thorough quantification of their overlap in a phenotype-specific manner remains elusive. Here, we have quantified the overlap of genes identified through large-scale genome-wide association studies (GWASs) for 62 complex traits and diseases with genes containing mutations known to cause 20 broad categories of Mendelian disorders. We identified a significant enrichment of genes linked to phenotypically matched Mendelian disorders in GWAS gene sets; of the total 1,240 comparisons, a higher proportion of phenotypically matched or related pairs (n = 50 of 92 [54%]) than phenotypically unmatched pairs (n = 27 of 1,148 [2%]) demonstrated significant overlap, confirming a phenotype-specific enrichment pattern. Further, we observed elevated GWAS effect sizes near genes linked to phenotypically matched Mendelian disorders. Finally, we report examples of GWAS variants localized at the transcription start site or physically interacting with the promoters of genes linked to phenotypically matched Mendelian disorders. Our results are consistent with the hypothesis that genes that are disrupted in Mendelian disorders are dysregulated by non-coding variants in complex traits and demonstrate how leveraging findings from related Mendelian disorders and functional genomic datasets can prioritize genes that are putatively dysregulated by local and distal non-coding GWAS variants.
Collapse
|