1
|
Abu-Arja MH, Brown AL, Su JM, Okcu MF, Lindsay HB, McGovern SL, McAleer MF, Grosshans DR, Chintagumpala MM, Paulino AC. The cochlear dose and the age at radiotherapy predict severe hearing loss after passive scattering proton therapy and cisplatin in children with medulloblastoma. Neuro Oncol 2024; 26:1912-1920. [PMID: 38916058 PMCID: PMC11449093 DOI: 10.1093/neuonc/noae114] [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: 03/25/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Hearing loss (HL) is associated with worse neurocognitive outcomes among patients with medulloblastoma. We aimed to identify risk factors associated with severe HL and to evaluate the generalizability of a published HL calculator among patients treated with passive scattering proton therapy (PSPT) and cisplatin. METHODS We identified patients aged 3-21 years who were treated at our centers between 2007 and 2022. Audiograms were graded using the International Society of Pediatric Oncology (SIOP) Boston scale. Time to grades 3-4 HL was evaluated using Kaplan-Meier and multivariable Cox models to estimate hazard ratios and 95% confidence intervals (CI). RESULTS Seventy-nine patients were treated with PSPT at a median age of 7.5 years (range: 3.1-21.1). The mean cochlear dose (Dmc) (±SD) was 31.5 ± 8.5 Gy, and the cumulative cisplatin dose was 295 ± 50 mg/m2. Fifty-nine patients (75%) received amifostine. Patients completed a median of 9 audiograms (range: 4-22) with a median audiogram follow-up of 49 months (range: 6-177). Twenty-seven patients (34%) had grades 3-4 HL. In adjusted Cox models, only higher Dmc (HR = 1.12, 95% CI:1.06-1.18) was associated with grades 3-4 HL. The predicted 3-year incidence of grades 3-4 HL was 40.0% (95% CI: 21.3-66.3) and 66.7% (95% CI: 35.4-93.7) for children with Dmc ≥36 Gy and age at radiotherapy ≥7 and <7 years, respectively (P = .042). It was 8.9% (95% CI: 2.3-31.6) and 15.6% (95% CI: 5.3-41.1) for children with Dmc <36 Gy and age at radiotherapy ≥7 and <7 years, respectively (P = .78). CONCLUSIONS Children <7 years at radiotherapy with a Dmc ≥36 Gy are at higher risk for HL.
Collapse
Affiliation(s)
- Mohammad H Abu-Arja
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Austin L Brown
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Jack M Su
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - M Fatih Okcu
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Holly B Lindsay
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Susan L McGovern
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mary Frances McAleer
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David R Grosshans
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Murali M Chintagumpala
- Division of Hematology/Oncology, Department of Pediatrics, Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Arnold C Paulino
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Wu J, Wu Y, Zhao T, Wang X, Guo Q, Wang S, Chen S, Ju X, Li J, Wu X, Zheng Z. Targeting RAC1 reactivates pyroptosis to reverse paclitaxel resistance in ovarian cancer by suppressing P21-activated kinase 4. MedComm (Beijing) 2024; 5:e719. [PMID: 39224538 PMCID: PMC11366825 DOI: 10.1002/mco2.719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Pyroptosis may play an important role in the resistance of ovarian cancer (OC) to chemotherapy. However, the mechanism by which pyroptosis modulation can attenuate chemotherapy resistance has not been comprehensively studied in OC. Here, we demonstrated that RAS-associated C3 botulinum toxin substrate 1 (RAC1) is highly expressed in OC and is negatively correlated with patient outcomes. Through cell function tests and in vivo tumor formation tests, we found that RAC1 can promote tumor growth by mediating paclitaxel (PTX) resistance. RAC1 can mediate OC progression by inhibiting pyroptosis, as evidenced by high-throughput automated confocal imaging, the release of lactate dehydrogenase (LDH), the expression of the inflammatory cytokines IL-1β/IL-18 and the nucleotide oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. Mechanically, RNA-seq, gene set enrichment analysis (GSEA), coimmunoprecipitation (Co-IP), mass spectrometry (MS), and ubiquitination tests further confirmed that RAC1 inhibits caspase-1/gasdermin D (GSDMD)-mediated canonical pyroptosis through the P21-activated kinase 4 (PAK4)/mitogen-activated protein kinase (MAPK) pathway, thereby promoting PTX resistance in OC cells. Finally, the whole molecular pathway was verified by the results of in vivo drug combination tests, clinical specimen detection and the prognosis. In summary, our results suggest that the combination of RAC1 inhibitors with PTX can reverse PTX resistance by inducing pyroptosis through the PAK4/MAPK pathway.
Collapse
Affiliation(s)
- Jiangchun Wu
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yong Wu
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Tianyi Zhao
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiangwei Wang
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghaiChina
| | - Qinhao Guo
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Simin Wang
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Siyu Chen
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xingzhu Ju
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jin Li
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiaohua Wu
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhong Zheng
- Department of Gynaecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
3
|
Patil V, Perez-Carpena P, Lopez-Escamez JA. A systematic review on the contribution of DNA methylation to hearing loss. Clin Epigenetics 2024; 16:88. [PMID: 38970134 PMCID: PMC11227199 DOI: 10.1186/s13148-024-01697-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/17/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND DNA methylation may have a regulatory role in monogenic sensorineural hearing loss and complex, polygenic phenotypic forms of hearing loss, including age-related hearing impairment or Meniere disease. The purpose of this systematic review is to critically assess the evidence supporting a functional role of DNA methylation in phenotypes associated with hearing loss. RESULTS The search strategy yielded a total of 661 articles. After quality assessment, 25 records were selected (12 human DNA methylation studies, 5 experimental animal studies and 8 studies reporting mutations in the DNMT1 gene). Although some methylation studies reported significant differences in CpG methylation in diverse gene promoters associated with complex hearing loss phenotypes (ARHI, otosclerosis, MD), only one study included a replication cohort that supported a regulatory role for CpG methylation in the genes TCF25 and POLE in ARHI. Conversely, several studies have independently confirmed pathogenic mutations within exon 21 of the DNMT1 gene, which encodes the DNA (cytosine-5)-methyltransferase 1 enzyme. This methylation enzyme is strongly associated with a rare disease defined by autosomal dominant cerebellar ataxia, deafness and narcolepsy (ADCA-DN). Of note, rare variants in DNMT1 and DNMT3A genes have also been reported in noise-induced hearing loss. CONCLUSIONS Evidence supporting a functional role for DNA methylation in hearing loss is limited to few genes in complex disorders such as ARHI. Mutations in the DNMT1 gene are associated with ADCA-DN, suggesting the CpG methylation in hearing loss genes deserves further attention in hearing research.
Collapse
Affiliation(s)
- Vibha Patil
- Meniere's Disease Neuroscience Research Program, Faculty of Medicine and Health, School of Medical Sciences, The Kolling Institute, University of Sydney, Rm 611024, Level 11 Kolling Institute | 10 Westbourne St, St Leonards, Sydney, NSW, 2064, Australia.
| | - Patricia Perez-Carpena
- Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, Ibs.Granada, Universidad de Granada, Granada, Spain
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitaria, ibs.GRANADA, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Program, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
- Department of Otolaryngology, Hospital Universitario San Cecilio, Instituto de Investigacion Biosanitaria, ibs.GRANADA, Granada, Spain
| | - Jose A Lopez-Escamez
- Meniere's Disease Neuroscience Research Program, Faculty of Medicine and Health, School of Medical Sciences, The Kolling Institute, University of Sydney, Rm 611024, Level 11 Kolling Institute | 10 Westbourne St, St Leonards, Sydney, NSW, 2064, Australia
- Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, Ibs.Granada, Universidad de Granada, Granada, Spain
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitaria, ibs.GRANADA, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Program, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| |
Collapse
|
4
|
Sayer M, Ng DQ, Chan R, Kober K, Chan A. Current evidence supporting associations of DNA methylation measurements with survivorship burdens in cancer survivors: A scoping review. Cancer Med 2024; 13:e7470. [PMID: 38963018 PMCID: PMC11222976 DOI: 10.1002/cam4.7470] [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: 01/17/2024] [Revised: 05/27/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
INTRODUCTION Identifying reliable biomarkers that reflect cancer survivorship symptoms remains a challenge for researchers. DNA methylation (DNAm) measurements reflecting epigenetic changes caused by anti-cancer therapy may provide needed insights. Given lack of consensus describing utilization of DNAm data to predict survivorship issues, a review evaluating the current landscape is warranted. OBJECTIVE Provide an overview of current studies examining associations of DNAm with survivorship burdens in cancer survivors. METHODS A literature review was conducted including studies if they focused on cohorts of cancer survivors, utilized peripheral blood cell DNAm data, and evaluated the associations of DNAm and survivorship issues. RESULTS A total of 22 studies were identified, with majority focused on breast (n = 7) or childhood cancer (n = 9) survivors, and half studies included less than 100 patients (n = 11). Survivorship issues evaluated included those related to neurocognition (n = 5), psychiatric health (n = 3), general wellness (n = 9), chronic conditions (n = 5), and treatment specific toxicities (n = 4). Studies evaluated epigenetic age metrics (n = 10) and DNAm levels at individual CpG sites or regions (n = 12) for their associations with survivorship issues in cancer survivors along with relevant confounding factors. Significant associations of measured DNAm in the peripheral blood samples of cancer survivors and survivorship issues were identified. DISCUSSION/CONCLUSION Studies utilizing epigenetic age metrics and differential methylation analysis demonstrated significant associations of DNAm measurements with survivorship burdens. Associations were observed encompassing diverse survivorship outcomes and timeframes relative to anti-cancer therapy initiation. These findings underscore the potential of these measurements as useful biomarkers in survivorship care and research.
Collapse
Affiliation(s)
- Michael Sayer
- School of Pharmacy and Pharmaceutical SciencesUniversity of California IrvineIrvineCaliforniaUSA
| | - Ding Quan Ng
- School of Pharmacy and Pharmaceutical SciencesUniversity of California IrvineIrvineCaliforniaUSA
| | - Raymond Chan
- School of Nursing and Health SciencesFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Kord Kober
- School of NursingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Alexandre Chan
- School of Pharmacy and Pharmaceutical SciencesUniversity of California IrvineIrvineCaliforniaUSA
| |
Collapse
|
5
|
Murphy B, Jackson A, Bass JK, Tsang DS, Ronckers CM, Kremer L, Baliga S, Olch A, Zureick AH, Jee KW, Constine LS, Yock TI. Modeling the Risk of Hearing Loss From Radiation Therapy in Childhood Cancer Survivors: A PENTEC Comprehensive Review. Int J Radiat Oncol Biol Phys 2024; 119:446-456. [PMID: 37855793 DOI: 10.1016/j.ijrobp.2023.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/28/2023] [Accepted: 08/06/2023] [Indexed: 10/20/2023]
Abstract
PURPOSE The Pediatric Normal Tissue Effects in the Clinic (PENTEC) hearing loss (HL) task force reviewed investigations on cochlear radiation dose-response relationships and risk factors for developing HL. Evidence-based dose-response data are quantified to guide treatment planning. METHODS AND MATERIALS A systematic review of the literature was performed to correlate HL with cochlear dosimetry. HL was considered present if a threshold exceeded 20 dB at any frequency. Radiation dose, ototoxic chemotherapy exposure, hearing profile including frequency spectra, interval to HL, and age at radiation therapy (RT) were analyzed. RESULTS Literature was systematically reviewed from 1970 to 2021. This resulted in 739 abstracts; 19 met inclusion for meta-analysis, and 4 included data amenable to statistical modeling. These 4 studies included 457 cochleas at risk in patients treated with RT without chemotherapy, and 398 cochlea treated with chemotherapy. The incidence and severity of cochlear HL from RT exposure alone is related to dose and age. Risk of HL was <5% in cochlea receiving a mean dose ≤35 Gy but increased to 30% at 50 Gy. HL risk ranged from 25% to 40% in children under the age of 5 years at diagnosis, declining to 10% in older children for any radiation dose. Probability of similar severe HL occurred at doses 18.3 Gy higher for children <3 versus >3 years of age. High-frequency HL was most common, with average onset occurring 3.6 years (range, 0.4-13.2 years) after RT. Exposure to platinum-based chemotherapies added to the rates of HL at a given cochlear dose level, with 300 mg/m2 shifting the dose response by 7 Gy. CONCLUSIONS In children treated with RT alone, risk of HL was low for cochlear dose <35 Gy and rose when dose exceeded 35 Gy without clear RT dose dependence. High-frequency HL was most prevalent, but all frequencies were affected. Children younger than 5 years were at highest risk of developing HL, although independent effects of dose and age were not fully elucidated. Future reports with more granular data are needed to better delineate time to onset of HL and the effects of chemoradiotherapy.
Collapse
Affiliation(s)
- Blair Murphy
- Department of Radiation Medicine, Oregon Health & Science University, Doernbecher Children's Hospital, Portland, Oregon.
| | - Andrew Jackson
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Johnnie K Bass
- Rehabilitation Services, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Cecile M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Pediatric Oncology, Emma Children's Hospital/Academic Medical Center, Amsterdam, The Netherlands; Division of Childhood Cancer Epidemiology, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Leontien Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Emma Children's Hospital, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
| | - Sujith Baliga
- Ohio State University Medical Center, Columbus, Ohio
| | - Arthur Olch
- University of Southern California, Children's Hospital of Los Angeles, Los Angeles, California
| | | | | | - Louis S Constine
- Departments of Radiation Oncology and Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Torunn I Yock
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Lupo PJ, Petrick LM, Hoang TT, Janitz AE, Marcotte EL, Schraw JM, Arora M, Scheurer ME. Using primary teeth and archived dried spots for exposomic studies in children: Exploring new paths in the environmental epidemiology of pediatric cancer. Bioessays 2021; 43:e2100030. [PMID: 34106479 DOI: 10.1002/bies.202100030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022]
Abstract
It is estimated that 300,000 children 0-14 years of age are diagnosed with cancer worldwide each year. While the absolute risk of cancer in children is low, it is the leading cause of death due to disease in children in high-income countries. In spite of this, the etiologies of pediatric cancer are largely unknown. Environmental exposures have long been thought to play an etiologic role. However, to date, there are few well-established environmental risk factors for pediatric malignancies, likely due to technical barriers in collecting biological samples prospectively in pediatric populations for direct measurements. In this review, we propose the use of novel or underutilized biospecimens (dried blood spots and teeth) and molecular approaches for exposure assessment (epigenetics, metabolomics, and somatic mutational profiles). Future epidemiologic studies of pediatric cancer should incorporate novel exposure assessment methodologies, data on molecular features of tumors, and a more complete assessment of gene-environment interactions.
Collapse
Affiliation(s)
- Philip J Lupo
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Lauren M Petrick
- The Senator Frank R. Lautenberg Environmental Health Science Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thanh T Hoang
- Epidemiology Branch, National Institutes of Health, Department of Health and Human Services, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Amanda E Janitz
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Erin L Marcotte
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jeremy M Schraw
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Manish Arora
- The Senator Frank R. Lautenberg Environmental Health Science Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael E Scheurer
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
7
|
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
|
8
|
Role of epigenetic mechanisms in cisplatin-induced toxicity. Crit Rev Oncol Hematol 2019; 137:131-142. [PMID: 31014509 DOI: 10.1016/j.critrevonc.2019.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/13/2019] [Accepted: 03/13/2019] [Indexed: 12/16/2022] Open
Abstract
Cisplatin (CDDP) is a highly effective antineoplastic agent, widely used in the treatment of various malignant tumors. However, its major problems are side effects associated to toxicity. Considerable inter-individual differences have been reported for CDDP-induced toxicity due to genetic and epigenetic factors. Genetic causes are well described; however, epigenetic modifications are not fully addressed. In the last few years, many evidences were found linking microRNA to the development of CDDP-mediated toxicity, particularly nephrotoxicity. In this review, we described how genetic and epigenetic modifications can be important determinants for the development of toxicity in patients treated with CDDP, and how these alterations may be interesting biomarkers for monitoring toxicity induced by CDDP. Considering the validation in different studies, we suggest that miR-34a, -146b, -378a, -192, and -193 represent an attractive study group to evaluate potential biomarkers to detect CDDP-related nephrotoxicity.
Collapse
|
9
|
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
|
10
|
Ness KK, Kirkland JL, Gramatges MM, Wang Z, Kundu M, McCastlain K, Li-Harms X, Zhang J, Tchkonia T, Pluijm SMF, Armstrong GT. Premature Physiologic Aging as a Paradigm for Understanding Increased Risk of Adverse Health Across the Lifespan of Survivors of Childhood Cancer. J Clin Oncol 2018; 36:2206-2215. [PMID: 29874132 DOI: 10.1200/jco.2017.76.7467] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The improvement in survival of childhood cancer observed across the past 50 years has resulted in a growing acknowledgment that simply extending the lifespan of survivors is not enough. It is incumbent on both the cancer research and the clinical care communities to also improve the health span of survivors. It is well established that aging adult survivors of childhood cancer are at increased risk of chronic health conditions, relative to the general population. However, as the first generation of survivors age into their 50s and 60s, it has become increasingly evident that this population is also at risk of early onset of physiologic aging. Geriatric measures have uncovered evidence of reduced strength and speed and increased fatigue, all components of frailty, among survivors with a median age of 33 years, which is similar to adults older than 65 years of age in the general population. Furthermore, frailty in survivors independently increased the risk of morbidity and mortality. Although there has been a paucity of research investigating the underlying biologic mechanisms for advanced physiologic age in survivors, results from geriatric populations suggest five biologically plausible mechanisms that may be potentiated by exposure to cancer therapies: increased cellular senescence, reduced telomere length, epigenetic modifications, somatic mutations, and mitochondrial DNA infidelity. There is now a critical need for research to elucidate the biologic mechanisms of premature aging in survivors of childhood cancer. This research could pave the way for new frontiers in the prevention of these life-changing outcomes.
Collapse
Affiliation(s)
- Kirsten K Ness
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - James L Kirkland
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Maria Monica Gramatges
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Zhaoming Wang
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mondira Kundu
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kelly McCastlain
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Xiujie Li-Harms
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jinghui Zhang
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Tamar Tchkonia
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Saskia Martine Francesca Pluijm
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Gregory T Armstrong
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|