1
|
Wu S, Xiong T, Guo S, Zhu C, He J, Wang S. An up-to-date view of paclitaxel-induced peripheral neuropathy. J Cancer Res Ther 2023; 19:1501-1508. [PMID: 38156915 DOI: 10.4103/jcrt.jcrt_1982_22] [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: 09/21/2022] [Accepted: 07/12/2023] [Indexed: 01/03/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN),referring to the damage to the peripheral nerves caused by exposure to a neurotoxic chemotherapeutic agent, is a common side effect amongst patients undergoing chemotherapy. Paclitaxel-induced peripheral neuropathy (PIPN) can lead to dose reduction or early cessation of chemotherapy, which is not conducive to patients'survival. Even after treatment is discontinued, PIPN symptoms carried a greater risk of worsening and plagued the patient's life, leading to long-term morbidity in survivors. Here, we summarize the research progress for clinical manifestations, risk factors, pathogenesis, prevention and treatment of PIPN, so as to embark on the path of preventing PIPN with prolongation of patient's life quality on a long-term basis.
Collapse
Affiliation(s)
- Shan Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pharmacy, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Tu Xiong
- Department of Radiology, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Shenglan Guo
- Department of Pharmacy, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Cuiyi Zhu
- Department of Pharmacy, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Jing He
- Department of Pharmacy, People's Hospital of Leshan, Shizhong, Leshan, China
| | - Shurong Wang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Pero ME, Chowdhury F, Bartolini F. Role of tubulin post-translational modifications in peripheral neuropathy. Exp Neurol 2023; 360:114274. [PMID: 36379274 PMCID: PMC11320756 DOI: 10.1016/j.expneurol.2022.114274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Peripheral neuropathy is a common disorder that results from nerve damage in the periphery. The degeneration of sensory axon terminals leads to changes or loss of sensory functions, often manifesting as debilitating pain, weakness, numbness, tingling, and disability. The pathogenesis of most peripheral neuropathies remains to be fully elucidated. Cumulative evidence from both early and recent studies indicates that tubulin damage may provide a common underlying mechanism of axonal injury in various peripheral neuropathies. In particular, tubulin post-translational modifications have been recently implicated in both toxic and inherited forms of peripheral neuropathy through regulation of axonal transport and mitochondria dynamics. This knowledge forms a new area of investigation with the potential for developing therapeutic strategies to prevent or delay peripheral neuropathy by restoring tubulin homeostasis.
Collapse
Affiliation(s)
- Maria Elena Pero
- Department of Pathology and Cell Biology, Columbia University, New York, USA; Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Italy
| | - Farihah Chowdhury
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
3
|
Kang L, Zhao Q, Jiang K, Yu X, Chao H, Yin L, Wang Y. Uncovering potential diagnostic biomarkers of acute myocardial infarction based on machine learning and analyzing its relationship with immune cells. BMC Cardiovasc Disord 2023; 23:2. [PMID: 36600215 DOI: 10.1186/s12872-022-02999-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/07/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a common cardiovascular disease. This study aimed to mine biomarkers associated with AMI to aid in clinical diagnosis and management. METHODS All mRNA and miRNA data were downloaded from public database. Differentially expressed mRNAs (DEmRNAs) and differentially expressed miRNAs (DEmiRNAs) were identified using the metaMA and limma packages, respectively. Functional analysis of the DEmRNAs was performed. In order to explore the relationship between miRNA and mRNA, we construct miRNA-mRNA negative regulatory network. Potential biomarkers were identified based on machine learning. Subsequently, ROC and immune correlation analysis were performed on the identified key DEmRNA biomarkers. RESULTS According to the false discovery rate < 0.05, 92 DEmRNAs and 272 DEmiRNAs were identified. GSEA analysis found that kegg_peroxisome was up-regulated in AMI and kegg_steroid_hormone_biosynthesis was down-regulated in AMI compared to normal controls. 5 key DEmRNA biomarkers were identified based on machine learning, and classification diagnostic models were constructed. The random forests (RF) model has the highest accuracy. This indicates that RF model has high diagnostic value and may contribute to the early diagnosis of AMI. ROC analysis found that the area under curve of 5 key DEmRNA biomarkers were all greater than 0.7. Pearson correlation analysis showed that 5 key DEmRNA biomarkers were correlated with most of the differential infiltrating immune cells. CONCLUSION The identification of new molecular biomarkers provides potential research directions for exploring the molecular mechanism of AMI. Furthermore, it is important to explore new diagnostic genetic biomarkers for the diagnosis and treatment of AMI.
Collapse
Affiliation(s)
- Ling Kang
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China
| | - Qiang Zhao
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China
| | - Ke Jiang
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China.
| | - Xiaoyan Yu
- Coronary Care Unit, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China
| | - Hui Chao
- Coronary Care Unit, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China
| | - Lijuan Yin
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China
| | - Yueqing Wang
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, No. 706, Taishan Street, Taian, 271000, Shandong, China.
| |
Collapse
|
4
|
Guijosa A, Freyria A, Espinosa‐Fernandez JR, Estrada‐Mena FJ, Armenta‐Quiroga AS, Ortega‐Treviño MF, Catalán R, Antonio‐Aguirre B, Villarreal‐Garza C, Perez‐Ortiz AC. Pharmacogenetics of taxane-induced neurotoxicity in breast cancer: Systematic review and meta-analysis. Clin Transl Sci 2022; 15:2403-2436. [PMID: 35892315 PMCID: PMC9579387 DOI: 10.1111/cts.13370] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/09/2022] [Accepted: 06/20/2022] [Indexed: 01/25/2023] Open
Abstract
Taxane-based chemotherapy regimens are used as first-line treatment for breast cancer. Neurotoxicity, mainly taxane-induced peripheral neuropathy (TIPN), remains the most important dose-limiting adverse event. Multiple genes may be associated with TIPN; however, the strength and direction of the association remain unclear. For this reason, we systematically reviewed observational studies of TIPN pharmacogenetic markers in breast cancer treatment. We conducted a systematic search of terms alluding to breast cancer, genetic markers, taxanes, and neurotoxicity in Ovid, ProQuest, PubMed, Scopus, Virtual Health, and Web of Science. We assessed the quality of evidence and bias profile. We extracted relevant variables and effect measures. Whenever possible, we performed random-effects gene meta-analyses and examined interstudy heterogeneity with meta-regression models and subgroup analyses. This study follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) and STrengthening the REporting of Genetic Association Studies (STREGA) reporting guidance. A total of 42 studies with 19,431 participants were included. These evaluated 262 single-nucleotide polymorphisms (SNPs) across 121 genes. We conducted meta-analyses on 23 genes with 60 SNPs (19 studies and 6246 participants). Thirteen individual SNPs (ABCB1-rs2032582, ABCB1-rs3213619, BCL6/-rs1903216, /CAND1-rs17781082, CYP1B1-rs1056836, CYP2C8-rs10509681, CYP2C8-rs11572080, EPHA5-rs7349683, EPHA6-rs301927, FZD3-rs7001034, GSTP1-rs1138272, TUBB2A-rs9501929, and XKR4-rs4737264) and the overall SNPs' effect in four genes (CYP3A4, EphA5, GSTP1, and SLCO1B1) were statistically significantly associated with TIPN through meta-analysis. In conclusion, through systematic review and meta-analysis, we found that polymorphisms, and particularly 13 SNPs, are associated with TIPN, suggesting that genetics does play a role in interindividual predisposition. Further studies could potentially use these findings to develop individual risk profiles and guide decision making.
Collapse
Affiliation(s)
| | - Ana Freyria
- School of MedicineUniversidad PanamericanaMexico CityMexico
| | | | | | | | | | - Rodrigo Catalán
- School of MedicineUniversidad PanamericanaMexico CityMexico,Thoracic Oncology UnitInstituto Nacional de CancerologíaMexico CityMexico
| | | | - Cynthia Villarreal‐Garza
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de MonterreySan Pedro Garza GarcíaNuevo LeónMexico
| | - Andric C. Perez‐Ortiz
- School of MedicineUniversidad PanamericanaMexico CityMexico,Transplant CenterMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
5
|
Shi X, Jiang X, Chen C, Zhang Y, Sun X. The interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases: Implications for therapy. Pharmacol Res 2022; 184:106452. [PMID: 36116706 DOI: 10.1016/j.phrs.2022.106452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Microtubules, a highly dynamic cytoskeleton, participate in many cellular activities including mechanical support, organelles interactions, and intracellular trafficking. Microtubule organization can be regulated by modification of tubulin subunits, microtubule-associated proteins (MAPs) or agents modulating microtubule assembly. Increasing studies demonstrate that microtubule disorganization correlates with various cardiocerebrovascular diseases including heart failure and ischemic stroke. Microtubules also mediate intracellular transport as well as intercellular transfer of mitochondria, a power house in cells which produce ATP for various physiological activities such as cardiac mechanical function. It is known to all that both microtubules and mitochondria participate in the progression of cancer and Parkinson's disease. However, the interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases remain unclear. In this paper, we will focus on the roles of microtubules in cardiocerebrovascular diseases, and discuss the interplay of mitochondria and microtubules in disease development and treatment. Elucidation of these issues might provide significant diagnostic value as well as potential targets for cardiocerebrovascular diseases.
Collapse
Affiliation(s)
- Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| | - Xuan Jiang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Congwei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
6
|
Chua KC, El-Haj N, Priotti J, Kroetz DL. Mechanistic insights into the pathogenesis of microtubule-targeting agent-induced peripheral neuropathy from pharmacogenetic and functional studies. Basic Clin Pharmacol Toxicol 2022; 130 Suppl 1:60-74. [PMID: 34481421 PMCID: PMC8716520 DOI: 10.1111/bcpt.13654] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/04/2021] [Accepted: 09/01/2021] [Indexed: 01/03/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting toxicity that affects 30%-40% of patients undergoing cancer treatment. Although multiple mechanisms of chemotherapy-induced neurotoxicity have been described in preclinical models, these have not been translated into widely effective strategies for the prevention or treatment of CIPN. Predictive biomarkers to inform therapeutic approaches are also lacking. Recent studies have examined genetic risk factors associated with CIPN susceptibility. This review provides an overview of the clinical and pathologic features of CIPN and summarizes efforts to identify target pathways through genetic and functional studies. Structurally and mechanistically diverse chemotherapeutics are associated with CIPN; however, the current review is focused on microtubule-targeting agents since these are the focus of most pharmacogenetic association and functional studies of CIPN. Genome-wide pharmacogenetic association studies are useful tools to identify not only causative genes and genetic variants but also genetic networks implicated in drug response or toxicity and have been increasingly applied to investigations of CIPN. Induced pluripotent stem cell-derived models of human sensory neurons are especially useful to understand the mechanistic significance of genomic findings. Combined genetic and functional genomic efforts to understand CIPN hold great promise for developing therapeutic approaches for its prevention and treatment.
Collapse
Affiliation(s)
- Katherina C. Chua
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California San Francisco, San Francisco, CA 94143-2911,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911
| | - Nura El-Haj
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911
| | - Josefina Priotti
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911
| | - Deanna L. Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143-2911
| |
Collapse
|
7
|
Cunningham GM, Shen F, Wu X, Cantor EL, Gardner L, Philips S, Jiang G, Bales CL, Tan Z, Liu Y, Wan J, Fehrenbacher JC, Schneider BP. The impact of SBF2 on taxane-induced peripheral neuropathy. PLoS Genet 2022; 18:e1009968. [PMID: 34986146 PMCID: PMC8765656 DOI: 10.1371/journal.pgen.1009968] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/18/2022] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Taxane-induced peripheral neuropathy (TIPN) is a devastating survivorship issue for many cancer patients. In addition to its impact on quality of life, this toxicity may lead to dose reductions or treatment discontinuation, adversely impacting survival outcomes and leading to health disparities in African Americans (AA). Our lab has previously identified deleterious mutations in SET-Binding Factor 2 (SBF2) that significantly associated with severe TIPN in AA patients. Here, we demonstrate the impact of SBF2 on taxane-induced neuronal damage using an ex vivo model of SBF2 knockdown of induced pluripotent stem cell-derived sensory neurons. Knockdown of SBF2 exacerbated paclitaxel changes to cell viability and neurite outgrowth while attenuating paclitaxel-induced sodium current inhibition. Our studies identified paclitaxel-induced expression changes specific to mature sensory neurons and revealed candidate genes involved in the exacerbation of paclitaxel-induced phenotypes accompanying SBF2 knockdown. Overall, these findings provide ex vivo support for the impact of SBF2 on the development of TIPN and shed light on the potential pathways involved.
Collapse
Affiliation(s)
- Geneva M. Cunningham
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Fei Shen
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Xi Wu
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Erica L. Cantor
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Laura Gardner
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Santosh Philips
- Department of Clinical Pharmacology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Casey L. Bales
- Department of Clinical Pharmacology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Zhiyong Tan
- Department of Pharmacology and Toxicology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Jill C. Fehrenbacher
- Department of Pharmacology and Toxicology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Bryan P. Schneider
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| |
Collapse
|
8
|
Tymon-Rosario J, Adjei NN, Roque DM, Santin AD. Microtubule-Interfering Drugs: Current and Future Roles in Epithelial Ovarian Cancer Treatment. Cancers (Basel) 2021; 13:6239. [PMID: 34944858 PMCID: PMC8699494 DOI: 10.3390/cancers13246239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Taxanes and epothilones are chemotherapeutic agents that ultimately lead to cell death through inhibition of normal microtubular function. This review summarizes the literature demonstrating their current use and potential promise as therapeutic agents in the treatment of epithelial ovarian cancer (EOC), as well as putative mechanisms of resistance. Historically, taxanes have become the standard of care in the front-line and recurrent treatment of epithelial ovarian cancer. In the past few years, epothilones (i.e., ixabepilone) have become of interest as they may retain activity in taxane-treated patients since they harbor several features that may overcome mechanisms of taxane resistance. Clinical data now support the use of ixabepilone in the treatment of platinum-resistant or refractory ovarian cancer. Clinical data strongly support the use of microtubule-interfering drugs alone or in combination in the treatment of epithelial ovarian cancer. Ongoing clinical trials will shed further light into the potential of making these drugs part of current standard practice.
Collapse
Affiliation(s)
- Joan Tymon-Rosario
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (J.T.-R.); (N.N.A.)
| | - Naomi N. Adjei
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (J.T.-R.); (N.N.A.)
| | - Dana M. Roque
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Alessandro D. Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (J.T.-R.); (N.N.A.)
| |
Collapse
|
9
|
Abstract
Chemotherapy-induced peripheral neurotoxicity (CIPN) is a major dose-limiting side effect of many anti-cancer agents, including taxanes, platinums, vinca alkaloids, proteasome inhibitors, immunomodulatory drugs, and antibody-drug conjugates. The resultant symptoms often persist post treatment completion and continue to impact on long-term function and quality of life for cancer survivors. At present, dose reduction remains the only strategy to prevent severe neuropathy, often leading clinicians to the difficult decision of balancing maximal treatment exposure and minimal long-lasting side effects. This review examines the clinical presentations of CIPN with each class of neurotoxic treatment, describing signs, symptoms, and long-term outcomes. We provide an update on the proposed mechanisms of nerve damage and review current data on clinical and genetic risk factors contributing to CIPN development. We also examine recent areas of research in the treatment and prevention of CIPN, with specific focus on current clinical trials and consensus recommendations for CIPN management.
Collapse
|
10
|
Garofano K, Park CS, Alarcon C, Avitia J, Barbour A, Diemert D, Fraser CM, Friedman PN, Horvath A, Rashid K, Shaazuddin M, Sidahmed A, O'Brien TJ, Perera MA, Lee NH. Differences in the Platelet mRNA Landscape Portend Racial Disparities in Platelet Function and Suggest Novel Therapeutic Targets. Clin Pharmacol Ther 2021; 110:702-713. [PMID: 34255863 DOI: 10.1002/cpt.2363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/07/2021] [Indexed: 11/10/2022]
Abstract
The African American (AA) population displays a 1.6 to 3-fold higher incidence of thrombosis and stroke mortality compared with European Americans (EAs). Current antiplatelet therapies target the ADP-mediated signaling pathway, which displays significant pharmacogenetic variation for platelet reactivity. The focus of this study was to define underlying population differences in platelet function in an effort to identify novel molecular targets for future antiplatelet therapy. We performed deep coverage RNA-Seq to compare gene expression levels in platelets derived from a cohort of healthy volunteers defined by ancestry determination. We identified > 13,000 expressed platelet genes of which 480 were significantly differentially expressed genes (DEGs) between AAs and EAs. DEGs encoding proteins known or predicted to modulate platelet aggregation, morphology, or platelet count were upregulated in AA platelets. Numerous G-protein coupled receptors, ion channels, and pro-inflammatory cytokines not previously associated with platelet function were likewise differentially expressed. Many of the signaling proteins represent potential pharmacologic targets of intervention. Notably, we confirmed the differential expression of cytokines IL32 and PROK2 in an independent cohort by quantitative real-time polymerase chain reaction, and provide functional validation of the opposing actions of these two cytokines on collagen-induced AA platelet aggregation. Using Genotype-Tissue Expression whole blood data, we identified 516 expression quantitative trait locuses with Fst values > 0.25, suggesting that population-differentiated alleles may contribute to differences in gene expression. This study identifies gene expression differences at the population level that may affect platelet function and serve as potential biomarkers to identify cardiovascular disease risk. Additionally, our analysis uncovers candidate novel druggable targets for future antiplatelet therapies.
Collapse
Affiliation(s)
- Kaitlin Garofano
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - C Sehwan Park
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cristina Alarcon
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Juan Avitia
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - April Barbour
- Department of Medicine, George Washington University, Washington, DC, USA
| | - David Diemert
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Claire M Fraser
- Institute for Genome Sciences, University of Maryland, Baltimore, Maryland, USA
| | - Paula N Friedman
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, USA
| | - Kameron Rashid
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Mohammed Shaazuddin
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alfateh Sidahmed
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Travis J O'Brien
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Minoli A Perera
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Norman H Lee
- Department of Pharmacology and Physiology, GW Cancer Center, George Washington University, Washington, DC, USA
| |
Collapse
|
11
|
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating “dying back” neuropathy featuring a distal-to-proximal peripheral nerve degeneration seen in cancer patients undergoing chemotherapy. The pathogenenic mechanisms of CIPN are largely unknown. We report that in sensory neurons, the CIPN-inducing drug bortezomib caused axonopathy and disrupted mitochondria motility by increasing delta 2 tubulin (D2), the only irreversible tubulin posttranslational modification and a marker of hyper-stable microtubules. These data provide a new paradigm for the risk associated with enhanced tubulin longevity in peripheral neuropathy and suggest that targeting the enzymes regulating this tubulin modification may provide therapies that prevent the axonal injury observed in bortezomib-induced peripheral neuropathy. The pathogenesis of chemotherapy-induced peripheral neuropathy (CIPN) is poorly understood. Here, we report that the CIPN-causing drug bortezomib (Bort) promotes delta 2 tubulin (D2) accumulation while affecting microtubule stability and dynamics in sensory neurons in vitro and in vivo and that the accumulation of D2 is predominant in unmyelinated fibers and a hallmark of bortezomib-induced peripheral neuropathy (BIPN) in humans. Furthermore, while D2 overexpression was sufficient to cause axonopathy and inhibit mitochondria motility, reduction of D2 levels alleviated both axonal degeneration and the loss of mitochondria motility induced by Bort. Together, our data demonstrate that Bort, a compound structurally unrelated to tubulin poisons, affects the tubulin cytoskeleton in sensory neurons in vitro, in vivo, and in human tissue, indicating that the pathogenic mechanisms of seemingly unrelated CIPN drugs may converge on tubulin damage. The results reveal a previously unrecognized pathogenic role for D2 in BIPN that may occur through altered regulation of mitochondria motility.
Collapse
|
12
|
Adjei AA, Lopez CL, Schaid DJ, Sloan JA, Le-Rademacher JG, Loprinzi CL, Norman AD, Olson JE, Couch FJ, Beutler AS, Vachon CM, Ruddy KJ. Genetic Predictors of Chemotherapy-Induced Peripheral Neuropathy from Paclitaxel, Carboplatin and Oxaliplatin: NCCTG/Alliance N08C1, N08CA and N08CB Study. Cancers (Basel) 2021; 13:1084. [PMID: 33802509 PMCID: PMC7959452 DOI: 10.3390/cancers13051084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common and potentially permanent adverse effect of chemotherapeutic agents including taxanes such as paclitaxel and platinum-based compounds such as oxaliplatin and carboplatin. Previous studies have suggested that genetics may impact the risk of CIPN. We conducted genome-wide association studies (GWASs) for CIPN in two independent populations who had completed European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ)-CIPN20 assessments (a CIPN-specific 20-item questionnaire which includes three scales that evaluate sensory, autonomic, and motor symptoms). The study population N08Cx included 692 participants from three clinical trials (North Central Cancer Treatment Group (NCCTG) N08C1, N08CA, and N08CB) who had been treated with paclitaxel, paclitaxel plus carboplatin, or oxaliplatin. The primary endpoint for the GWAS was the change from pre-chemotherapy CIPN20 sensory score to the worse score over the following 18 weeks. Study population The Mayo Clinic Breast Disease Registry (MCBDR) consisted of 381 Mayo Clinic Breast Disease Registry enrollees who had been treated with taxane or platinum-based chemotherapy. The primary endpoint for the GWAS assessed was the earliest CIPN20 sensory score available after the completion of chemotherapy. In multivariate model analyses, chemotherapy regimen (p = 3.0 × 10-8) and genetic ancestry (p = 0.007) were significantly associated with CIPN in the N08Cx population. Only age (p = 0.0004) was significantly associated with CIPN in the MCBDR population. The SNP most associated with CIPN was rs56360211 near PDE6C (p =7.92 × 10-8) in N08Cx and rs113807868 near TMEM150C in the MCBDR (p = 1.27 × 10-8). Due to a lack of replication, we cannot conclude that we identified any genetic predictors of CIPN.
Collapse
Affiliation(s)
- Araba A. Adjei
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (A.A.A.); (C.L.L.); (A.S.B.)
- Alliance Cancer Control Program, Mayo Clinic, Rochester, MN 55905, USA
| | - Camden L. Lopez
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Daniel J. Schaid
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Jeff A. Sloan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer G. Le-Rademacher
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles L. Loprinzi
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (A.A.A.); (C.L.L.); (A.S.B.)
- Alliance Cancer Control Program, Mayo Clinic, Rochester, MN 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Aaron D. Norman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Janet E. Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Andreas S. Beutler
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (A.A.A.); (C.L.L.); (A.S.B.)
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Celine M. Vachon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (C.L.L.); (D.J.S.); (J.A.S.); (J.G.L.-R.); (J.E.O.); (C.M.V.); (A.D.N.)
| | - Kathryn J. Ruddy
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (A.A.A.); (C.L.L.); (A.S.B.)
- Alliance Cancer Control Program, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
13
|
Hertz DL. Exploring pharmacogenetics of paclitaxel- and docetaxel-induced peripheral neuropathy by evaluating the direct pharmacogenetic-pharmacokinetic and pharmacokinetic-neuropathy relationships. Expert Opin Drug Metab Toxicol 2021; 17:227-239. [PMID: 33401943 DOI: 10.1080/17425255.2021.1856367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Peripheral neuropathy (PN) is an adverse effect of several classes of chemotherapy including the taxanes. Predictive PN biomarkers could inform individualized taxane treatment to reduce PN and enhance therapeutic outcomes. Pharmacogenetics studies of taxane-induced PN have focused on genes involved in pharmacokinetics, including enzymes and transporters. Contradictory findings from these studies prevent translation of genetic biomarkers into clinical practice. Areas covered: This review discusses the progress toward identifying pharmacogenetic predictors of PN by assessing the evidence for two independent associations; the effect of pharmacogenetics on taxane pharmacokinetics and the evidence that taxane pharmacokinetics affects PN. Assessing these direct relationships allows the reader to understand the progress toward individualized taxane treatment and future research opportunities. Expert opinion: Paclitaxel pharmacokinetics is a major determinant of PN. Additional clinical trials are needed to confirm the clinical benefit of individualized dosing to achieve target paclitaxel exposure. Genetics does not meaningfully contribute to paclitaxel pharmacokinetics and may not be useful to inform dosing. However, genetics may contribute to PN sensitivity and could be useful for estimating patients' optimal paclitaxel exposure. For docetaxel, genetics has not been demonstrated to have a meaningful effect on pharmacokinetics and there is no evidence that pharmacokinetics determines PN.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy , Ann Arbor, MI, United States
| |
Collapse
|
14
|
Khodzhaev K, Ng OH, Tugcu D, Erbilgin Y, Ng YY, Celkan T, Timur C, Karakas Z, Ozdemir GN, Yıldırmak Y, Sayitoglu M. High
TUBB2A
expression in childhood T‐ALL is correlated with the clinical outcome. Int J Lab Hematol 2020. [DOI: 10.1111/ijlh.13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Khusan Khodzhaev
- Genetics Department Aziz Sancar Institute of Experimental Medicine Istanbul University Istanbul Turkey
- Istanbul University Institute of Health Sciences Istanbul Turkey
| | - Ozden Hatirnaz Ng
- Department of Medical Biology School of Medicine Acibadem Mehmet Ali Aydinlar University Istanbul Turkey
| | - Deniz Tugcu
- Pediatric Hematology Oncology Department Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Yucel Erbilgin
- Genetics Department Aziz Sancar Institute of Experimental Medicine Istanbul University Istanbul Turkey
| | - Yuk Yin Ng
- Genetics and Bioengineering Department Istanbul Bilgi University Istanbul Turkey
| | - Tiraje Celkan
- Pediatric Hematology Oncology Department Istanbul University‐Cerrahpasa Faculty of Medicine Istanbul Turkey
| | - Cetin Timur
- Pediatric Hematology Oncology Department Seven Hill Hospital Istanbul Turkey
| | - Zeynep Karakas
- Pediatric Hematology Oncology Department Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Gul Nihal Ozdemir
- Pediatric Hematology Oncology Department Istanbul University‐Cerrahpasa Faculty of Medicine Istanbul Turkey
- Pediatric Hematology Division Istanbul Kanuni Sultan Suleyman Education and Research Hospital Istanbul Turkey
| | - Yıldız Yıldırmak
- Pediatric Hematology Division Ministry of Health Sisli Etfal Education and Research Hospital Istanbul Turkey
| | - Muge Sayitoglu
- Genetics Department Aziz Sancar Institute of Experimental Medicine Istanbul University Istanbul Turkey
| |
Collapse
|
15
|
Neurotoxicity of antineoplastic drugs: Mechanisms, susceptibility, and neuroprotective strategies. Adv Med Sci 2020; 65:265-285. [PMID: 32361484 DOI: 10.1016/j.advms.2020.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 12/22/2019] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
This review summarizes the adverse effects on the central and/or peripheral nervous systems that may occur in response to antineoplastic drugs. In particular, we describe the neurotoxic side effects of the most commonly used drugs, such as platinum compounds, doxorubicin, ifosfamide, 5-fluorouracil, vinca alkaloids, taxanes, methotrexate, bortezomib and thalidomide. Neurotoxicity may result from direct action of compounds on the nervous system or from metabolic alterations produced indirectly by these drugs, and either the central nervous system or the peripheral nervous system, or both, may be affected. The incidence and severity of neurotoxicity are principally related to the dose, to the duration of treatment, and to the dose intensity, though other factors, such as age, concurrent pathologies, and genetic predisposition may enhance the occurrence of side effects. To avoid or reduce the onset and severity of these neurotoxic effects, the use of neuroprotective compounds and/or strategies may be helpful, thereby enhancing the therapeutic effectiveness of antineoplastic drug.
Collapse
|
16
|
Plasma polymerized nanoparticles effectively deliver dual siRNA and drug therapy in vivo. Sci Rep 2020; 10:12836. [PMID: 32732927 PMCID: PMC7393381 DOI: 10.1038/s41598-020-69591-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
Multifunctional nanocarriers (MNCs) promise to improve therapeutic outcomes by combining multiple classes of molecules into a single nanostructure, enhancing active targeting of therapeutic agents and facilitating new combination therapies. However, nanocarrier platforms currently approved for clinical use can still only carry a single therapeutic agent. The complexity and escalating costs associated with the synthesis of more complex MNCs have been major technological roadblocks in the pathway for clinical translation. Here, we show that plasma polymerized nanoparticles (PPNs), synthesised in reactive gas discharges, can bind and effectively deliver multiple therapeutic cargo in a facile and cost-effective process compatible with up scaled commercial production. Delivery of siRNA against vascular endothelial growth factor (siVEGF) at extremely low concentrations (0.04 nM), significantly reduced VEGF expression in hard-to-transfect cells when compared with commercial platforms carrying higher siRNA doses (6.25 nM). PPNs carrying a combination of siVEGF and standard of care Paclitaxel (PPN-Dual) at reduced doses (< 100 µg/kg) synergistically modulated the microenvironment of orthotopic breast tumors in mice, and significantly reduced tumor growth. We propose PPNs as a new nanomaterial for delivery of therapeutics, which can be easily functionalised in any laboratory setting without the need for additional wet-chemistry and purification steps.
Collapse
|
17
|
Staff NP, Fehrenbacher JC, Caillaud M, Damaj MI, Segal RA, Rieger S. Pathogenesis of paclitaxel-induced peripheral neuropathy: A current review of in vitro and in vivo findings using rodent and human model systems. Exp Neurol 2020; 324:113121. [PMID: 31758983 PMCID: PMC6993945 DOI: 10.1016/j.expneurol.2019.113121] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/29/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022]
Abstract
Paclitaxel (Brand name Taxol) is widely used in the treatment of common cancers like breast, ovarian and lung cancer. Although highly effective in blocking tumor progression, paclitaxel also causes peripheral neuropathy as a side effect in 60-70% of chemotherapy patients. Recent efforts by numerous labs have aimed at defining the underlying mechanisms of paclitaxel-induced peripheral neuropathy (PIPN). In vitro models using rodent dorsal root ganglion neurons, human induced pluripotent stem cells, and rodent in vivo models have revealed a number of molecular pathways affected by paclitaxel within axons of sensory neurons and within other cell types, such as the immune system and peripheral glia, as well skin. These studies revealed that paclitaxel induces altered calcium signaling, neuropeptide and growth factor release, mitochondrial damage and reactive oxygen species formation, and can activate ion channels that mediate responses to extracellular cues. Recent studies also suggest a role for the matrix-metalloproteinase 13 (MMP-13) in mediating neuropathy. These diverse changes may be secondary to paclitaxel-induced microtubule transport impairment. Human genetic studies, although still limited, also highlight the involvement of cytoskeletal changes in PIPN. Newly identified molecular targets resulting from these studies could provide the basis for the development of therapies with which to either prevent or reverse paclitaxel-induced peripheral neuropathy in chemotherapy patients.
Collapse
Affiliation(s)
- Nathan P Staff
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology and Toxicology, University School of Medicine, Indianapolis, IN 46202, USA
| | - Martial Caillaud
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, USA
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sandra Rieger
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA.
| |
Collapse
|
18
|
Zirpoli GR, McCann SE, Sucheston-Campbell LE, Hershman DL, Ciupak G, Davis W, Unger JM, Moore HCF, Stewart JA, Isaacs C, Hobday TJ, Salim M, Hortobagyi GN, Gralow JR, Budd GT, Albain KS, Ambrosone CB. Supplement Use and Chemotherapy-Induced Peripheral Neuropathy in a Cooperative Group Trial (S0221): The DELCaP Study. J Natl Cancer Inst 2019; 109:4098262. [PMID: 29546345 DOI: 10.1093/jnci/djx098] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/24/2017] [Indexed: 12/24/2022] Open
Abstract
Background Chemotherapy-induced peripheral neuropathy (CIPN) can interfere with daily function and quality of life, and there are no known preventive approaches. In a cohort of breast cancer patients receiving paclitaxel as part of a clinical trial (SWOG 0221), we examined the use of dietary supplements both before diagnosis and during treatment in relation to CIPN. Methods At registration to S0221, 1225 breast cancer patients completed questionnaires regarding the use of multivitamins and supplements before and at diagnosis. A second questionnaire at six months queried use during treatment. Supplement use was evaluated in relation to CIPN, assessed via the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE v. 3.0) and the self-reported Functional Assessment of Cancer Therapy/Gynecologic Oncology Group Neurotoxicity (FACT/GOG-Ntx) subscale. Odds ratios (ORs) and 95% confidence intervals (CIs) were computed with logistic regression for the CTCAE analyses and ordinal regression for the FACT/GOG-Ntx analyses. Results Multivitamin use before diagnosis was associated with reduced symptoms of CIPN (CTCAE-adjusted OR = 0.60, 95% CI = 0.42 to 0.87; FACT/GOG-Ntx-adjusted OR = 0.78, 95% CI = 0.61 to 1.00). Use during treatment was marginally inversely associated with CIPN (CTCAE-adjusted OR = 0.73, 95% CI = 0.49 to 1.08; FACT/GOG-Ntx-adjusted OR = 0.77, 95% CI = 0.60 to 0.99). Other supplement use, either before diagnosis or during treatment, was not statistically significantly associated with CIPN. Conclusions Multivitamin use may be associated with reduced risk of CIPN, although individual dietary supplement use did not appreciably affect risk. Multivitamin use could be a surrogate for other related behaviors that are the actual drivers of the association with reduced CIPN. Without prospective randomized trials of vitamin supplementation, recommendations for use or changes to clinical practice are clearly not warranted.
Collapse
Affiliation(s)
- Gary R Zirpoli
- Roswell Park Cancer Institute, Buffalo, NY.,Massachusetts General Hospital, Boston, MA
| | | | | | | | | | | | - Joseph M Unger
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | | | | | | | | | | | - Kathy S Albain
- Loyola University Chicago Stritch School of Medicine, Chicago, IL
| | | |
Collapse
|
19
|
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
|
20
|
Park SB, Kwok JB, Asher R, Lee CK, Beale P, Selle F, Friedlander M. Clinical and genetic predictors of paclitaxel neurotoxicity based on patient- versus clinician-reported incidence and severity of neurotoxicity in the ICON7 trial. Ann Oncol 2017; 28:2733-2740. [PMID: 29117336 DOI: 10.1093/annonc/mdx491] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting toxicity of paclitaxel, with no reliable method to identify at-risk patients. We investigated the incidence and risk factors including genetic polymorphisms associated with the development of CIPN based on clinician and patient reporting of neuropathic symptoms. PATIENTS AND METHODS Risk factors for the development of CIPN were examined in 454 patients treated with paclitaxel/carboplatin from the International Collaboration on Ovarian Neoplasms 7 (ICON7) trial. Neuropathy was graded by clinicians by standard adverse event reporting and by patients utilising OV28 questionnaire. Genetic risk factors were examined by selecting six single nucleotide polymorphisms in genes associated with microtubule function. Risk factors were assessed via dose-to-event cox regression models. RESULTS Grade >2 neuropathy was reported by clinicians in 28% of patients, while 67% of patients reported 'quite a bit' or 'very much' tingling or numbness. Agreement between clinicians and patients was poor (κ = 0.236, 95% confidence interval, 0.177-0.296, P < 0.001). Older age, bevacizumab treatment and bowel resection were associated with clinician reported CIPN, while older age and volume of residual disease were associated with patient-reported neuropathy. There were no significant associations between clinician-reported neuropathy or patient-reported neuropathy and TUBB2, CEP72 or individual MAPT or GSK3B SNPs, however MAPT additive polymorphisms were associated with patient-reported neuropathy and GSK3B additive polymorphisms were associated with clinician reported CIPN. CONCLUSIONS There was significant discordance between patient- and clinician-reported neurotoxicity. The lack of consensus regarding optimal outcome measures and whose opinion with regard to CIPN takes precedence is a limitation in the investigation of risk factors for CIPN. Care must be taken to select and include patient-reported outcome measures in CIPN assessment to enable accurate identification of genetic and other risk factors for neuropathy.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/complications
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/complications
- Adenocarcinoma, Mucinous/drug therapy
- Adenocarcinoma, Mucinous/pathology
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents, Phytogenic/adverse effects
- Biomarkers, Tumor/genetics
- Cystadenocarcinoma, Serous/complications
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/pathology
- Endometrial Neoplasms/complications
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/pathology
- Female
- Follow-Up Studies
- Humans
- Incidence
- Middle Aged
- Neoplasm Invasiveness
- Neurotoxicity Syndromes/diagnosis
- Neurotoxicity Syndromes/epidemiology
- Neurotoxicity Syndromes/etiology
- Neurotoxicity Syndromes/genetics
- Outcome Assessment, Health Care
- Ovarian Neoplasms/complications
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Paclitaxel/adverse effects
- Patient Reported Outcome Measures
- Physicians
- Polymorphism, Single Nucleotide
- Prognosis
- Risk Factors
- Severity of Illness Index
- Surveys and Questionnaires
- Survival Rate
- Young Adult
Collapse
Affiliation(s)
- S B Park
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Sydney; Prince of Wales Clinical School
| | - J B Kwok
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Sydney; Neuroscience Research Australia, University of New South Wales, Sydney
| | - R Asher
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney
| | - C K Lee
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney
| | - P Beale
- Concord Cancer Centre, Concord Repatriation General Hospital, Sydney; Sydney Medical School, University of Sydney, Sydney, Australia
| | - F Selle
- Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens and Department of Medical Oncology, Alliance For Cancer Research, Hôpital Tenon, Paris, France
| | | |
Collapse
|
21
|
Cliff J, Jorgensen AL, Lord R, Azam F, Cossar L, Carr DF, Pirmohamed M. The molecular genetics of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2017; 120:127-140. [PMID: 29198326 DOI: 10.1016/j.critrevonc.2017.09.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/12/2017] [Accepted: 09/11/2017] [Indexed: 01/13/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) can adversely affect completion of systemic anti-cancer treatment and cause long-term morbidity. Increasingly pharmacogenetic studies have been performed to explore susceptibility to this important adverse effect. A systematic review was conducted to identify pharmacogenetic studies, assess their quality and findings and undertake meta-analysis where possible. 93 studies were included. Notable methodological issues included lack of standardisation and detail in phenotype definition and acknowledgement of potential confounding factors. Insufficient data was presented in many studies meaning only a minority could be included in meta-analysis showing mainly non-significant effects. Nonetheless, SNPs in CYP2C8, CYP3A4, ARHGEF10, EPHA and TUBB2A genes (taxanes), FARS2, ACYP2 and TAC1 (oxaliplatin), and CEP75 and CYP3A5 (vincristine) are of potential interest. These require exploration in large cohort studies with robust methodology and well-defined phenotypes. Seeking standardisation of phenotype, collaboration and subsequently, individual-patient-data meta-analysis may facilitate identifying contributory SNPs which could be combined in a polygenic risk score to predict those most at risk of CIPN.
Collapse
Affiliation(s)
- J Cliff
- University of Liverpool, Liverpool, L69 3BX, UK; Clatterbridge Cancer Centre NHS Foundation Trust, Clatterbridge Road, Wirral, CH63 4JY, UK.
| | | | - R Lord
- University of Liverpool, Liverpool, L69 3BX, UK; Clatterbridge Cancer Centre NHS Foundation Trust, Clatterbridge Road, Wirral, CH63 4JY, UK.
| | - F Azam
- Clatterbridge Cancer Centre NHS Foundation Trust, Clatterbridge Road, Wirral, CH63 4JY, UK.
| | - L Cossar
- University of Liverpool, Liverpool, L69 3BX, UK; Clatterbridge Cancer Centre NHS Foundation Trust, Clatterbridge Road, Wirral, CH63 4JY, UK.
| | - D F Carr
- University of Liverpool, Liverpool, L69 3BX, UK.
| | - M Pirmohamed
- University of Liverpool, Liverpool, L69 3BX, UK.
| |
Collapse
|
22
|
Fukuda Y, Li Y, Segal RA. A Mechanistic Understanding of Axon Degeneration in Chemotherapy-Induced Peripheral Neuropathy. Front Neurosci 2017; 11:481. [PMID: 28912674 PMCID: PMC5583221 DOI: 10.3389/fnins.2017.00481] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Chemotherapeutic agents cause many short and long term toxic side effects to peripheral nervous system (PNS) that drastically alter quality of life. Chemotherapy-induced peripheral neuropathy (CIPN) is a common and enduring disorder caused by several anti-neoplastic agents. CIPN typically presents with neuropathic pain, numbness of distal extremities, and/or oversensitivity to thermal or mechanical stimuli. This adverse side effect often requires a reduction in chemotherapy dosage or even discontinuation of treatment. Currently there are no effective treatment options for CIPN. While the underlying mechanisms for CIPN are not understood, current data identify a “dying back” axon degeneration of distal nerve endings as the major pathology in this disorder. Therefore, mechanistic understanding of axon degeneration will provide insights into the pathway and molecular players responsible for CIPN. Here, we review recent findings that expand our understanding of the pathogenesis of CIPN and discuss pathways that may be shared with the axonal degeneration that occurs during developmental axon pruning and during injury-induced Wallerian degeneration. These mechanistic insights provide new avenues for development of therapies to prevent or treat CIPN.
Collapse
Affiliation(s)
- Yusuke Fukuda
- Department of Neurobiology, Harvard Medical SchoolBoston, MA, United States.,Department of Cancer Biology, Dana-Farber Cancer InstituteBoston, MA, United States
| | - Yihang Li
- Department of Neurobiology, Harvard Medical SchoolBoston, MA, United States.,Department of Cancer Biology, Dana-Farber Cancer InstituteBoston, MA, United States
| | - Rosalind A Segal
- Department of Neurobiology, Harvard Medical SchoolBoston, MA, United States.,Department of Cancer Biology, Dana-Farber Cancer InstituteBoston, MA, United States
| |
Collapse
|
23
|
Taxanes and platinum derivatives impair Schwann cells via distinct mechanisms. Sci Rep 2017; 7:5947. [PMID: 28729624 PMCID: PMC5519765 DOI: 10.1038/s41598-017-05784-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/05/2017] [Indexed: 12/31/2022] Open
Abstract
Impairment of peripheral neurons by anti-cancer agents, including taxanes and platinum derivatives, has been considered to be a major cause of chemotherapy-induced peripheral neuropathy (CIPN), however, the precise underlying mechanisms are not fully understood. Here, we examined the direct effects of anti-cancer agents on Schwann cells. Exposure of primary cultured rat Schwann cells to paclitaxel (0.01 μM), cisplatin (1 μM), or oxaliplatin (3 μM) for 48 h induced cytotoxicity and reduced myelin basic protein expression at concentrations lower than those required to induce neurotoxicity in cultured rat dorsal root ganglion (DRG) neurons. Similarly, these anti-cancer drugs disrupted myelin formation in Schwann cell/DRG neuron co-cultures without affecting nerve axons. Cisplatin and oxaliplatin, but not paclitaxel, caused mitochondrial dysfunction in cultured Schwann cells. By contrast, paclitaxel led to dedifferentiation of Schwann cells into an immature state, characterized by increased expression of p75 and galectin-3. Consistent with in vitro findings, repeated injection of paclitaxel increased expression of p75 and galectin-3 in Schwann cells within the mouse sciatic nerve. These results suggest that taxanes and platinum derivatives impair Schwan cells by inducing dedifferentiation and mitochondrial dysfunction, respectively, which may be important in the development of CIPN in conjunction with their direct impairment in peripheral neurons.
Collapse
|
24
|
Staff NP, Grisold A, Grisold W, Windebank AJ. Chemotherapy-induced peripheral neuropathy: A current review. Ann Neurol 2017; 81:772-781. [PMID: 28486769 PMCID: PMC5656281 DOI: 10.1002/ana.24951] [Citation(s) in RCA: 481] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/30/2017] [Accepted: 05/01/2017] [Indexed: 12/16/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting side effect experienced by patients receiving treatment for cancer. Approximately 30 to 40% of patients treated with neurotoxic chemotherapy will develop CIPN, and there is considerable variability in its severity between patients. It is often sensory-predominant with pain and can lead to long-term morbidity in survivors. The prevalence and burden of CIPN late effects will likely increase as cancer survival rates continue to improve. In this review, we discuss the approach to peripheral neuropathy in patients with cancer and address the clinical phenotypes and pathomechanisms of specific neurotoxic chemotherapeutic agents. Ann Neurol 2017;81:772-781.
Collapse
Affiliation(s)
| | - Anna Grisold
- Department of Neurology, Medical University of Vienna, Austria
| | - Wolfgang Grisold
- Ludwig Boltzmann Institute for Experimental und Clinical
Traumatology, Vienna, Austria
| | | |
Collapse
|
25
|
Starobova H, Vetter I. Pathophysiology of Chemotherapy-Induced Peripheral Neuropathy. Front Mol Neurosci 2017; 10:174. [PMID: 28620280 PMCID: PMC5450696 DOI: 10.3389/fnmol.2017.00174] [Citation(s) in RCA: 380] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/17/2017] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy-induced neuropathy is a common, dose-dependent adverse effect of several antineoplastics. It can lead to detrimental dose reductions and discontinuation of treatment, and severely affects the quality of life of cancer survivors. Clinically, chemotherapy-induced peripheral neuropathy presents as deficits in sensory, motor, and autonomic function which develop in a glove and stocking distribution due to preferential effects on longer axons. The pathophysiological processes are multi-factorial and involve oxidative stress, apoptotic mechanisms, altered calcium homeostasis, axon degeneration and membrane remodeling as well as immune processes and neuroinflammation. This review focusses on the commonly used antineoplastic substances oxaliplatin, cisplatin, vincristine, docetaxel, and paclitaxel which interfere with the cancer cell cycle-leading to cell death and tumor degradation-and cause severe acute and chronic peripheral neuropathies. We discuss drug mechanism of action and pharmacokinetic disposition relevant to the development of peripheral neuropathy, the epidemiology and clinical presentation of chemotherapy-induced neuropathy, emerging insight into genetic susceptibilities as well as current understanding of the pathophysiology and treatment approaches.
Collapse
Affiliation(s)
- Hana Starobova
- Centre for Pain Research, Institute for Molecular Bioscience, University of QueenslandSt Lucia, QLD, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, University of QueenslandSt Lucia, QLD, Australia.,School of Pharmacy, University of QueenslandSt Lucia, QLD, Australia
| |
Collapse
|
26
|
Schneider BP, Lai D, Shen F, Jiang G, Radovich M, Li L, Gardner L, Miller KD, O'Neill A, Sparano JA, Xue G, Foroud T, Sledge GW. Charcot-Marie-Tooth gene, SBF2, associated with taxane-induced peripheral neuropathy in African Americans. Oncotarget 2016; 7:82244-82253. [PMID: 27732968 PMCID: PMC5347688 DOI: 10.18632/oncotarget.12545] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/18/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Taxane-induced peripheral neuropathy (TIPN) is one of the most important survivorship issues for cancer patients. African Americans (AA) have previously been shown to have an increased risk for this toxicity. Germline predictive biomarkers were evaluated to help identify a priori which patients might be at extraordinarily high risk for this toxicity. EXPERIMENTAL DESIGN Whole exome sequencing was performed using germline DNA from 213 AA patients who received a standard dose and schedule of paclitaxel in the adjuvant, randomized phase III breast cancer trial, E5103. Cases were defined as those with either grade 3-4 (n=64) or grade 2-4 (n=151) TIPN and were compared to controls (n=62) that were not reported to have experienced TIPN. We retained for analysis rare variants with a minor allele frequency <3% and which were predicted to be deleterious by protein prediction programs. A gene-based, case-control analysis using SKAT was performed to identify genes that harbored an imbalance of deleterious variants associated with increased risk of TIPN. RESULTS Five genes had a p-value < 10-4 for grade 3-4 TIPN analysis and three genes had a p-value < 10-4 for the grade 2-4 TIPN analysis. For the grade 3-4 TIPN analysis, SET binding factor 2 (SBF2) was significantly associated with TIPN (p-value=4.35 x10-6). Five variants were predicted to be deleterious in SBF2. Inherited mutations in SBF2 have previously been associated with autosomal recessive, Type 4B2 Charcot-Marie-Tooth (CMT) disease. CONCLUSION Rare variants in SBF2, a CMT gene, predict an increased risk of TIPN in AA patients receiving paclitaxel.
Collapse
Affiliation(s)
| | - Dongbing Lai
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Fei Shen
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Guanglong Jiang
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Milan Radovich
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lang Li
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Laura Gardner
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kathy D. Miller
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Anne O'Neill
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts, USA
| | - Joseph A. Sparano
- Albert Einstein University, Montefiore Medical Center, Bronx, New York, USA
| | - Gloria Xue
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tatiana Foroud
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - George W. Sledge
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
27
|
Lam SW, Frederiks CN, van der Straaten T, Honkoop AH, Guchelaar HJ, Boven E. Genotypes of CYP2C8 and FGD4 and their association with peripheral neuropathy or early dose reduction in paclitaxel-treated breast cancer patients. Br J Cancer 2016; 115:1335-1342. [PMID: 27736846 PMCID: PMC5129817 DOI: 10.1038/bjc.2016.326] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 12/16/2022] Open
Abstract
Background: The purpose of this study was to evaluate single-nucleotide polymorphisms (SNPs) in genes encoding key metabolising enzymes or involved in pharmacodynamics for possible associations with paclitaxel-induced peripheral neuropathy. Methods: The study population consists of 188 women from the multicenter, randomised, phase II ATX trial (BOOG2006-06; EudraCT number 2006-006058-83) that received paclitaxel and bevacizumab without or with capecitabine as first-line palliative therapy of HER2-negative metastatic breast cancer. Genotyping of CYP2C8*3 (c.416G>A), CYP3A4*22 (c.522-191C>T), TUBB2A (c.-101T>C), FGD4 (c.2044-236G>A) and EPHA5 (c.2895G>A) was performed by real-time PCR. Toxicity endpoints were cumulative dose (1) until first onset of grade ⩾1 peripheral neuropathy and (2) until first paclitaxel dose reduction from related toxicity (NCI-CTCAE version 3.0). SNPs were evaluated using the Kaplan–Meier method, the Gehan–Breslow–Wilcoxon test and the multivariate Cox regression analysis. Results: The rate of grade ⩾1 peripheral neuropathy was 67% (n=126). The rate of dose reduction was 46% (n=87). Age ⩾65 years was a risk factor for peripheral neuropathy (HR=1.87, P<0.008), but not for dose reduction. When adjusted for age, body surface area and total cumulative paclitaxel dose, CYP2C8*3 carriers had an increased risk of peripheral neuropathy (HR=1.59, P=0.045). FGD4 c.2044-236 A-allele carriers had an increased risk of paclitaxel dose reduction (HR per A-allele=1.38, P=0.036) when adjusted for total cumulative paclitaxel dose. Conclusions: These findings may point towards clinically useful indicators of early toxicity, but warrant further investigation.
Collapse
Affiliation(s)
- Siu W Lam
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Charlotte N Frederiks
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tahar van der Straaten
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aafke H Honkoop
- Department of Medical Oncology, Isala Clinics, Zwolle, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Epie Boven
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
28
|
Apellániz-Ruiz M, Tejero H, Inglada-Pérez L, Sánchez-Barroso L, Gutiérrez-Gutiérrez G, Calvo I, Castelo B, Redondo A, García-Donás J, Romero-Laorden N, Sereno M, Merino M, Currás-Freixes M, Montero-Conde C, Mancikova V, Åvall-Lundqvist E, Green H, Al-Shahrour F, Cascón A, Robledo M, Rodríguez-Antona C. Targeted Sequencing Reveals Low-Frequency Variants in EPHA Genes as Markers of Paclitaxel-Induced Peripheral Neuropathy. Clin Cancer Res 2016; 23:1227-1235. [PMID: 27582484 DOI: 10.1158/1078-0432.ccr-16-0694] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/29/2016] [Accepted: 08/16/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Neuropathy is the dose-limiting toxicity of paclitaxel and a major cause for decreased quality of life. Genetic factors have been shown to contribute to paclitaxel neuropathy susceptibility; however, the major causes for interindividual differences remain unexplained. In this study, we identified genetic markers associated with paclitaxel-induced neuropathy through massive sequencing of candidate genes.Experimental Design: We sequenced the coding region of 4 EPHA genes, 5 genes involved in paclitaxel pharmacokinetics, and 30 Charcot-Marie-Tooth genes, in 228 cancer patients with no/low neuropathy or high-grade neuropathy during paclitaxel treatment. An independent validation series included 202 paclitaxel-treated patients. Variation-/gene-based analyses were used to compare variant frequencies among neuropathy groups, and Cox regression models were used to analyze neuropathy along treatment.Results: Gene-based analysis identified EPHA6 as the gene most significantly associated with paclitaxel-induced neuropathy. Low-frequency nonsynonymous variants in EPHA6 were present exclusively in patients with high neuropathy, and all affected the ligand-binding domain of the protein. Accumulated dose analysis in the discovery series showed a significantly higher neuropathy risk for EPHA5/6/8 low-frequency nonsynonymous variant carriers [HR, 14.60; 95% confidence interval (CI), 2.33-91.62; P = 0.0042], and an independent cohort confirmed an increased neuropathy risk (HR, 2.07; 95% CI, 1.14-3.77; P = 0.017). Combining the series gave an estimated 2.5-fold higher risk of neuropathy (95% CI, 1.46-4.31; P = 9.1 × 10-4).Conclusions: This first study sequencing EPHA genes revealed that low-frequency variants in EPHA6, EPHA5, and EPHA8 contribute to the susceptibility to paclitaxel-induced neuropathy. Furthermore, EPHA's neuronal injury repair function suggests that these genes might constitute important neuropathy markers for many neurotoxic drugs. Clin Cancer Res; 23(5); 1227-35. ©2016 AACR.
Collapse
Affiliation(s)
- María Apellániz-Ruiz
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Héctor Tejero
- Translational Bioinformatics Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Lucía Inglada-Pérez
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Lara Sánchez-Barroso
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Isabel Calvo
- Medical Oncology Department, Hospital Montepríncipe, Madrid, Spain.,Medical Oncology Department, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Beatriz Castelo
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Andrés Redondo
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Jesús García-Donás
- Gynecological and Genitourinary Tumors Programme, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Nuria Romero-Laorden
- Gynecological and Genitourinary Tumors Programme, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - María Sereno
- Medical Oncology Department, Hospital Universitario Infanta Sofía, Madrid, Spain
| | - María Merino
- Medical Oncology Department, Hospital Universitario Infanta Sofía, Madrid, Spain
| | - María Currás-Freixes
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristina Montero-Conde
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Veronika Mancikova
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elisabeth Åvall-Lundqvist
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköpings Universitet, Linköping, Sweden
| | - Henrik Green
- Clinical Pharmacology, Division of Drug Research, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköpings Universitet, Linköping, Sweden.,Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Fátima Al-Shahrour
- Translational Bioinformatics Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Cristina Rodríguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain. .,ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| |
Collapse
|
29
|
Juth V, Holman EA, Chan MK, Cramer SC. Genetics as a molecular window into recovery, its treatment, and stress responses after stroke. J Investig Med 2016; 64:983-8. [PMID: 27045100 PMCID: PMC4942179 DOI: 10.1136/jim-2016-000126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2016] [Indexed: 01/13/2023]
Abstract
Stroke remains a major source of adult disability in the USA and worldwide. Most patients show some recovery during the weeks to months following a stroke, but this is generally incomplete. An emerging branch of therapeutics targets the processes underlying this behavioral recovery from stroke toward the goal of reducing long-term disability. A key factor hampering these efforts is the very large degree of variability between stroke survivors. Available data suggest that genetic differences could explain an important fraction of the differences between subjects. The current review considers this from several angles, including genetic differences in relation to drugs that promote recovery. Genetic factors related to physiological and psychological stress responses may also be critically important to understanding recovery after stroke and its treatment. The studies reviewed provide insights into recovery and suggest directions for further research to improve clinical decision-making in this setting. Genetic differences between patients might be used to help clinical trials select specific patient subgroups, on a biological basis, in order to sharpen the precision with which new treatments are evaluated. Pharmacogenomic factors might also provide insights into inter-subject differences in treatment side effects for pharmacological prescriptions, and behavioral interventions, and others. These efforts must be conducted with the strictest ethical standards given the highly sensitive nature of genetic data. Understanding the effect of selected genetic measures could improve a clinician's ability to predict the risk and efficacy of a restorative therapy and to make maximally informed decisions, and in so doing, facilitate individual patient care.
Collapse
Affiliation(s)
- Vanessa Juth
- Program in Nursing Science, UC Irvine, Irvine, California, USA
| | - E Alison Holman
- Program in Nursing Science, UC Irvine, Irvine, California, USA
| | - Michelle K Chan
- Program in Nursing Science, UC Irvine, Irvine, California, USA
| | | |
Collapse
|
30
|
Morrison G, Liu C, Wing C, Delaney SM, Zhang W, Dolan ME. Evaluation of inter-batch differences in stem-cell derived neurons. Stem Cell Res 2015; 16:140-8. [PMID: 26774046 DOI: 10.1016/j.scr.2015.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 01/24/2023] Open
Abstract
Differentiated cells retain the genetic information of the donor but the extent to which phenotypic differences between donors or batches of differentiated cells are explained by variation introduced during the differentiation process is not fully understood. In this study, we evaluated four separate batches of commercially available neurons originating from the same iPSCs to investigate whether the differentiation process used in manufacturing iPSCs to neurons affected genome-wide gene expression and modified cytosines, or neuronal sensitivity to drugs. No significant changes in gene expression, as measured by RNA-Seq, or cytosine modification levels, as measured by the Illumina 450K arrays, were observed between batches relative to changes over time. As expected, neurotoxic chemotherapeutics affected neuronal outgrowth, but no inter-batch differences were observed in sensitivity to paclitaxel, vincristine and cisplatin. As a testament to the utility of the model for studies of neuropathy, we observed that genes involved in neuropathy had relatively higher expression levels in these samples across different time points. Our results suggest that the process used to differentiate iPSCs into neurons is consistent, resulting in minimal intra-individual variability across batches. Therefore, this model is reasonable for studies of human neuropathy, druggable targets to prevent neuropathy, and other neurological diseases.
Collapse
Affiliation(s)
- Gladys Morrison
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, IL 60637, USA
| | - Cong Liu
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Claudia Wing
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Shannon M Delaney
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Wei Zhang
- Department of Preventive Medicine & The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - M Eileen Dolan
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, IL 60637, USA; Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
31
|
Frederiks C, Lam S, Guchelaar H, Boven E. Genetic polymorphisms and paclitaxel- or docetaxel-induced toxicities: A systematic review. Cancer Treat Rev 2015; 41:935-50. [DOI: 10.1016/j.ctrv.2015.10.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/17/2015] [Accepted: 10/20/2015] [Indexed: 12/28/2022]
|
32
|
Schneider BP, Li L, Radovich M, Shen F, Miller KD, Flockhart DA, Jiang G, Vance G, Gardner L, Vatta M, Bai S, Lai D, Koller D, Zhao F, O'Neill A, Smith ML, Railey E, White C, Partridge A, Sparano J, Davidson NE, Foroud T, Sledge GW. Genome-Wide Association Studies for Taxane-Induced Peripheral Neuropathy in ECOG-5103 and ECOG-1199. Clin Cancer Res 2015; 21:5082-5091. [PMID: 26138065 PMCID: PMC4717479 DOI: 10.1158/1078-0432.ccr-15-0586] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE Taxane-induced peripheral neuropathy (TIPN) is an important survivorship issue for many cancer patients. Currently, there are no clinically implemented biomarkers to predict which patients might be at increased risk for TIPN. We present a comprehensive approach to identification of genetic variants to predict TIPN. EXPERIMENTAL DESIGN We performed a genome-wide association study (GWAS) in 3,431 patients from the phase III adjuvant breast cancer trial, ECOG-5103 to compare genotypes with TIPN. We performed candidate validation of top SNPs for TIPN in another phase III adjuvant breast cancer trial, ECOG-1199. RESULTS When evaluating for grade 3-4 TIPN, 120 SNPs had a P value of <10(-4) from patients of European descent (EA) in ECOG-5103. Thirty candidate SNPs were subsequently tested in ECOG-1199 and SNP rs3125923 was found to be significantly associated with grade 3-4 TIPN (P = 1.7 × 10(-3); OR, 1.8). Race was also a major predictor of TIPN, with patients of African descent (AA) experiencing increased risk of grade 2-4 TIPN (HR, 2.1; P = 5.6 × 10(-16)) and grade 3-4 TIPN (HR, 2.6; P = 1.1 × 10(-11)) compared with others. An SNP in FCAMR, rs1856746, had a trend toward an association with grade 2-4 TIPN in AA patients from the GWAS in ECOG-5103 (OR, 5.5; P = 1.6 × 10(-7)). CONCLUSIONS rs3125923 represents a validated SNP to predict grade 3-4 TIPN. Genetically determined AA race represents the most significant predictor of TIPN.
Collapse
Affiliation(s)
| | - Lang Li
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Milan Radovich
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Fei Shen
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Kathy D. Miller
- Indiana University School of Medicine, Indianapolis,
Indiana
| | | | - Guanglong Jiang
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Gail Vance
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Laura Gardner
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Matteo Vatta
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Shaochun Bai
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Dongbing Lai
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Daniel Koller
- Indiana University School of Medicine, Indianapolis,
Indiana
| | - Fengmin Zhao
- Dana Farber Cancer Institute, Boston, Massachusetts
| | - Anne O'Neill
- Dana Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | - Joseph Sparano
- Albert Einstein university, Montefiore Medical Center,
Bronx, New York
| | | | - Tatiana Foroud
- Indiana University School of Medicine, Indianapolis,
Indiana
| | | |
Collapse
|
33
|
Brewer JR, Morrison G, Dolan ME, Fleming GF. Chemotherapy-induced peripheral neuropathy: Current status and progress. Gynecol Oncol 2015; 140:176-83. [PMID: 26556766 DOI: 10.1016/j.ygyno.2015.11.011] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 11/03/2015] [Accepted: 11/06/2015] [Indexed: 12/17/2022]
Abstract
As there are increasing numbers of cancer survivors, more attention is being paid to the long term unwanted effects patients may experience as a result of their treatment and the impact these side effects can have on their quality of life. Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most common long-term toxicities from chemotherapy. In this review we will briefly review the clinical presentation, evaluation and management of chemotherapy-induced peripheral neuropathy, with a focus on CIPN related to platinum and taxane agents. We will then discuss current clinical models of peripheral neuropathy and ongoing research to better understand CIPN and develop potential treatment options.
Collapse
Affiliation(s)
- Jamie R Brewer
- Section of Hematology-Oncology, Department of Medicine, University of Chicago Medical Center, 5841 S. Maryland Ave, Chicago, IL 60637, United States
| | - Gladys Morrison
- Section of Hematology-Oncology, Department of Medicine, The University of Chicago, Knapp Center for Biomedical Discovery, 900 East 57th Street, Chicago, IL 60637, United States
| | - M Eileen Dolan
- Section of Hematology-Oncology, Department of Medicine, The University of Chicago, Knapp Center for Biomedical Discovery, 900 East 57th Street, Chicago, IL 60637, United States
| | - Gini F Fleming
- Section of Hematology-Oncology, Department of Medicine, The University of Chicago, Knapp Center for Biomedical Discovery, 900 East 57th Street, Chicago, IL 60637, United States.
| |
Collapse
|
34
|
Srinivasan S, Clements JA, Batra J. Single nucleotide polymorphisms in clinics: Fantasy or reality for cancer? Crit Rev Clin Lab Sci 2015; 53:29-39. [DOI: 10.3109/10408363.2015.1075469] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
35
|
Reyes-Gibby CC, Wang J, Yeung SCJ, Shete S. Informative gene network for chemotherapy-induced peripheral neuropathy. BioData Min 2015; 8:24. [PMID: 26269716 PMCID: PMC4534051 DOI: 10.1186/s13040-015-0058-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 08/04/2015] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Host genetic variability has been implicated in chemotherapy-induced peripheral neuropathy (CIPN). A dose-limiting toxicity for chemotherapy agents, CIPN is also a debilitating condition that may progress to chronic neuropathic pain. We utilized a bioinformatics approach, which captures the complexity of intracellular and intercellular interactions, to identify genes for CIPN. METHODS Using genes pooled from the literature as a starting point, we used Ingenuity Pathway Analysis (IPA) to generate gene networks for CIPN. RESULTS We performed IPA core analysis for genes associated with platinum-, taxane- and platinum-taxane-induced neuropathy. We found that IL6, TNF, CXCL8, IL1B and ERK1/2 were the top genes in terms of the number of connections in platinum-induced neuropathy and TP53, MYC, PARP1, P38 MAPK and TNF for combined taxane-platinum-induced neuropathy. CONCLUSION Neurotoxicity is common in cancer patients treated with platinum compounds and anti-microtubule agents and CIPN is one of the debilitating sequela. The bioinformatic approach helped identify genes associated with CIPN in cancer patients.
Collapse
Affiliation(s)
- Cielito C. Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
36
|
Shimizu C. [Side effects of anticancer treatment and the needs for translational research on toxicity: a clinician's perspective]. Nihon Yakurigaku Zasshi 2015; 146:72-5. [PMID: 26256743 DOI: 10.1254/fpj.146.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
37
|
De Iuliis F, Taglieri L, Salerno G, Lanza R, Scarpa S. Taxane induced neuropathy in patients affected by breast cancer: Literature review. Crit Rev Oncol Hematol 2015; 96:34-45. [PMID: 26004917 DOI: 10.1016/j.critrevonc.2015.04.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/10/2015] [Accepted: 04/30/2015] [Indexed: 12/11/2022] Open
Abstract
Taxane induced neuropathy (TIN) is the most limiting side effect of taxane based chemotherapy, relative to the majority of breast cancer patients undergoing therapy with both docetaxel and paclitaxel. The symptoms begin symmetrically from the toes, because the tips of the longest nerves are affected for first. The patients report sensory symptoms such as paresthesia, dysesthesia, numbness, electric shock-like sensation, motor impairment and neuropathic pain. There is a great inter-individual variability among breast cancer women treated with taxanes, in fact 20-30% of them don't develop neurotoxicity. Actually, there is no standard therapy for TIN, although many medications, antioxidants and natural substances have been tested in vitro and in vivo. We will summarize all most recent literature data on TIN prevention and treatment, in order to reach an improvement in TIN management. Further studies are needed to evaluate new therapies that restore neuronal function and improve life quality of patients.
Collapse
Affiliation(s)
- Francesca De Iuliis
- Experimental Medicine Department, Sapienza University, viale Regina Elena 324, 00161 Rome, Italy
| | - Ludovica Taglieri
- Experimental Medicine Department, Sapienza University, viale Regina Elena 324, 00161 Rome, Italy
| | - Gerardo Salerno
- Experimental Medicine Department, Sapienza University, viale Regina Elena 324, 00161 Rome, Italy
| | - Rosina Lanza
- Ginecology and Obstetrics Department, Sapienza University, viale Regina Elena 324, 00161 Rome, Italy
| | - Susanna Scarpa
- Experimental Medicine Department, Sapienza University, viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
38
|
Modeling chemotherapeutic neurotoxicity with human induced pluripotent stem cell-derived neuronal cells. PLoS One 2015; 10:e0118020. [PMID: 25689802 PMCID: PMC4331516 DOI: 10.1371/journal.pone.0118020] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/04/2015] [Indexed: 12/14/2022] Open
Abstract
There are no effective agents to prevent or treat chemotherapy-induced peripheral neuropathy (CIPN), the most common non-hematologic toxicity of chemotherapy. Therefore, we sought to evaluate the utility of human neuron-like cells derived from induced pluripotent stem cells (iPSCs) as a means to study CIPN. We used high content imaging measurements of neurite outgrowth phenotypes to compare the changes that occur to iPSC-derived neuronal cells among drugs and among individuals in response to several classes of chemotherapeutics. Upon treatment of these neuronal cells with the neurotoxic drug paclitaxel, vincristine or cisplatin, we identified significant differences in five morphological phenotypes among drugs, including total outgrowth, mean/median/maximum process length, and mean outgrowth intensity (P < 0.05). The differences in damage among drugs reflect differences in their mechanisms of action and clinical CIPN manifestations. We show the potential of the model for gene perturbation studies by demonstrating decreased expression of TUBB2A results in significantly increased sensitivity of neurons to paclitaxel (0.23 ± 0.06 decrease in total neurite outgrowth, P = 0.011). The variance in several neurite outgrowth and apoptotic phenotypes upon treatment with one of the neurotoxic drugs is significantly greater between than within neurons derived from four different individuals (P < 0.05), demonstrating the potential of iPSC-derived neurons as a genetically diverse model for CIPN. The human neuron model will allow both for mechanistic studies of specific genes and genetic variants discovered in clinical studies and for screening of new drugs to prevent or treat CIPN.
Collapse
|
39
|
Park SB, Kwok JB, Loy CT, Friedlander ML, Lin CSY, Krishnan AV, Lewis CR, Kiernan MC. Paclitaxel-induced neuropathy: potential association of MAPT and GSK3B genotypes. BMC Cancer 2014; 14:993. [PMID: 25535399 PMCID: PMC4364586 DOI: 10.1186/1471-2407-14-993] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 12/16/2014] [Indexed: 02/07/2023] Open
Abstract
Background Paclitaxel treatment produces dose-limiting peripheral neurotoxicity, which adversely affects treatment and long-term outcomes. In the present study, the contribution of genetic polymorphisms to paclitaxel-induced neurotoxicity were assessed in 21 patients, focusing on polymorphisms involved in the tau-microtubule pathway, an important target of paclitaxel involved in neurotoxicity development. Methods Polymorphisms in the microtubule-associated protein tau (MAPT) gene (haplotype 1 and rs242557 polymorphism) and the glycogen synthase kinase-3β (GSK3β) gene (rs6438552 polymorphism) were investigated. Neurotoxicity was assessed using neuropathy grading scales, neurophysiological studies and patient questionnaires. Results A significant relationship between the GSK-3B rs6438552 polymorphism and paclitaxel-induced neurotoxicity was evident. Conclusions Polymorphisms in tau-associated genes may contribute to the development of paclitaxel-induced neurotoxicity, although larger series will be necessary to confirm these findings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Matthew C Kiernan
- Brain and Mind Research Institute, University of Sydney, Sydney, Australia.
| |
Collapse
|
40
|
Klute K, Nackos E, Tasaki S, Nguyen DP, Bander NH, Tagawa ST. Microtubule inhibitor-based antibody-drug conjugates for cancer therapy. Onco Targets Ther 2014; 7:2227-36. [PMID: 25506226 PMCID: PMC4259504 DOI: 10.2147/ott.s46887] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The specificity of monoclonal antibodies represents a potential therapeutic advantage, but their use as single agents in oncology has proven limited to date. The development of antibody-drug conjugates (ADCs) takes advantage of the specificity of the monoclonal antibody and potent cytotoxic effect of chemotherapy, leading to enhanced cytotoxicity in target cells and limiting toxicity to normal tissue. Microtubules represent a validated oncologic target in a range of tumor types, with a number of anti-microtubule targeting cytotoxic drugs approved for cancer use. The systemic use of potent microtubule-binding agents is limited by their effects in normal cells, which leads to toxicity including myelosuppression and peripheral neuropathy. Linking these agents to monoclonal antibodies may limit toxicity to normal tissues and increase drug concentration in target tissues, also allowing the use of more potent agents which would be too toxic to administer in their unbound form. Two such ADCs have been approved for clinical use and many others are in development. Here we review the characteristics of each of the ADC components that have led to efficacious therapies and discuss some of the tubulin inhibitor-based ADCs in development for cancer therapy.
Collapse
Affiliation(s)
- Kelsey Klute
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Eleni Nackos
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Shinsuke Tasaki
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Daniel P Nguyen
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Neil H Bander
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Scott T Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA ; Department of Urology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
41
|
Using Pharmacogene Polymorphism Panels to Detect Germline Pharmacodynamic Markers in Oncology. Clin Cancer Res 2014; 20:2530-40. [DOI: 10.1158/1078-0432.ccr-13-2780] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Integrated systems pharmacology analysis of clinical drug-induced peripheral neuropathy. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e114. [PMID: 24827872 PMCID: PMC4051377 DOI: 10.1038/psp.2014.11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/03/2014] [Indexed: 01/08/2023]
Abstract
A systems pharmacology approach was undertaken to define and identify the proteins/genes significantly associated with clinical incidence and severity of drug-induced peripheral neuropathy (DIPN). Pharmacological networks of 234 DIPN drugs, their known targets (both intended and unintended), and the intermediator proteins/genes interacting with these drugs via their known targets were examined. A permutation test identified 230 DIPN-associated intermediators that were enriched with apoptosis and stress response genes. Neuropathy incidence and severity were curated from drug labels and literature and were used to build a predictive model of DIPN using a regression tree algorithm, based on the drug targets and their intermediators. DIPN drugs whose targets interacted with both v-myc avian myelocytomatosis viral oncogene homolog (MYC) and proliferating cell nuclear antigen-associated factor (PAF15) were associated with a neuropathy incidence of 38.1%, whereas drugs interacting only with MYC had an incidence of 2.9%. These results warrant further investigation in order to develop a predictive tool for the DIPN potential of a new drug.
Collapse
|
43
|
Pearson-Fuhrhop KM, Cramer SC. Pharmacogenetics of neural injury recovery. Pharmacogenomics 2014; 14:1635-43. [PMID: 24088134 DOI: 10.2217/pgs.13.152] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Relatively few pharmacological agents are part of routine care for neural injury, although several are used or under consideration in acute stroke, chronic stroke, traumatic brain injury and secondary stroke prevention. Tissue plasminogen activator is approved for the treatment of acute ischemic stroke, and genetic variants may impact the efficacy and safety of this drug. In the chronic phase of stroke, several drugs such as L-dopa, fluoxetine and donepezil are under investigation for enhancing rehabilitation therapy, with varying levels of evidence. One potential reason for the mixed efficacy displayed by these drugs may be the influence of genetic factors that were not considered in prior studies. An understanding of the genetics impacting the efficacy of dopaminergic, serotonergic and cholinergic drugs may allow clinicians to target these potential therapies to those patients most likely to benefit. In the setting of stroke prevention, which is directly linked to neural injury recovery, the most highly studied pharmacogenomic interactions pertain to clopidogrel and warfarin. Incorporating pharmacogenomics into neural injury recovery has the potential to maximize the benefit of several current and potential pharmacological therapies and to refine the choice of pharmacological agent that may be used to enhance benefits from rehabilitation therapy.
Collapse
Affiliation(s)
- Kristin M Pearson-Fuhrhop
- Department of Anatomy & Neurobiology, University of California, Irvine, 200 S Manchester Avenue, Suite 206, Orange, CA 92868, USA
| | | |
Collapse
|
44
|
Abraham JE, Guo Q, Dorling L, Tyrer J, Ingle S, Hardy R, Vallier AL, Hiller L, Burns R, Jones L, Bowden SJ, Dunn JA, Poole CJ, Caldas C, Pharoah PPD, Earl HM. Replication of genetic polymorphisms reported to be associated with taxane-related sensory neuropathy in patients with early breast cancer treated with Paclitaxel. Clin Cancer Res 2014; 20:2466-75. [PMID: 24599932 DOI: 10.1158/1078-0432.ccr-13-3232] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Associations between taxane-related sensory neuropathy (TRSN) and single-nucleotide polymorphisms (SNP) have previously been reported, but few have been replicated in large, independent validation studies. This study evaluates the association between previously investigated SNPs and TRSN, using genotype data from a study of chemotherapy-related toxicity in patients with breast cancer. EXPERIMENTAL DESIGN We investigated 73 SNPs in 50 genes for their contribution to TRSN risk, using genotype data from 1,303 European patients. TRSN was assessed using National Cancer Institute common toxicity criteria for adverse events classification. Unconditional logistic regression evaluated the association between each SNP and TRSN risk (primary analysis). Cox regression analysis assessed the association between each SNP and cumulative taxane dose causing the first reported moderate/severe TRSN (secondary analysis). The admixture likelihood (AML) test, which considers all SNPs with a prior probability of association with TRSN together, tested the hypothesis that certain SNPs are truly associated. RESULTS The AML test provided strong evidence for the association of some SNPs with TRSN (P = 0.023). The two most significantly associated SNPs were rs3213619(ABCB1) [OR = 0.47; 95% confidence interval (CI), 0.28-0.79; P = 0.004] and rs9501929(TUBB2A) (OR = 1.80; 95% CI, 1.20-2.72; P = 0.005). A further 9 SNPs were significant at P-value ≤ 0.05. CONCLUSION This is currently the largest study investigating SNPs associated with TRSN. We found strong evidence that SNPs within genes in taxane pharmacokinetic and pharmacodynamic pathways contribute to TRSN risk. However, a large proportion of the inter-individual variability in TRSN remains unexplained. Further validated results from GWAS will help to identify new pathways, genes, and SNPs involved in TRSN susceptibility.
Collapse
Affiliation(s)
- Jean E Abraham
- Authors' Affiliations: Department of Oncology and Strangeways Research Laboratory, University of Cambridge; Cambridge Breast Unit and NIHR Cambridge Biomedical Research Centre, University of Cambridge NHS Foundation Hospitals; Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way; Cambridge Experimental Cancer Medicine Centre, Cambridge; Warwick Clinical Trials Unit, University of Warwick; and Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hertz DL, Roy S, Jack J, Motsinger-Reif AA, Drobish A, Clark LS, Carey LA, Dees EC, McLeod HL. Genetic heterogeneity beyond CYP2C8*3 does not explain differential sensitivity to paclitaxel-induced neuropathy. Breast Cancer Res Treat 2014; 145:245-54. [PMID: 24706167 DOI: 10.1007/s10549-014-2910-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/06/2014] [Indexed: 12/16/2022]
Abstract
The development of paclitaxel-induced peripheral neuropathy (PIPN) is influenced by drug exposure and patient genetics. The purpose of this analysis was to expand on a previous reported association of CYP2C8*3 and PIPN risk by investigating additional polymorphisms in CYP2C8 and in hundreds of other genes potentially relevant to paclitaxel pharmacokinetics. Clinical data was collected prospectively in an observational registry of newly diagnosed breast cancer patients. Patients treated with paclitaxel-containing regimens were genotyped using the Affymetrix DMET™ Plus chip. Patients who carried the CYP2C8*2, *3, or *4 variant were collapsed into a low-metabolizer CYP2C8 phenotype for association with PIPN. Separately, all SNPs that surpassed quality control were assessed individually and as a composite of genetic ancestry for associations with PIPN. 412 paclitaxel-treated patients and 564 genetic markers were included in the analysis. The risk of PIPN was significantly greater in the CYP2C8 low-metabolizer group (HR = 1.722, p = 0.018); however, the influences of the *2 and *4 SNPs were not independently significant (*2: p = 0.847, *4: p = 0.408). One intronic SNP in ABCG1 (rs492338) surpassed the exploratory significance threshold for an association with PIPN in the Caucasian cohort (p = 0.0008) but not in the non-Caucasian replication group (p = 0.54). Substantial genetic variability was observed within self-reported racial groups but this genetic variability was not associated with risk of grade 2+ PIPN. The pharmacogenetic heterogeneity within a cohort of breast cancer patients is dramatic, though we did not find evidence that this heterogeneity directly influences the risk of PIPN beyond the contribution of CYP2C8*3.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical, Social, and Administrative Sciences, University of Michigan College of Pharmacy, 428 Church St, Ann Arbor, MI, 48109, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cushion TD, Paciorkowski AR, Pilz DT, Mullins JGL, Seltzer LE, Marion RW, Tuttle E, Ghoneim D, Christian SL, Chung SK, Rees MI, Dobyns WB. De novo mutations in the beta-tubulin gene TUBB2A cause simplified gyral patterning and infantile-onset epilepsy. Am J Hum Genet 2014; 94:634-41. [PMID: 24702957 DOI: 10.1016/j.ajhg.2014.03.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 03/14/2014] [Indexed: 12/22/2022] Open
Abstract
Tubulins, and microtubule polymers into which they incorporate, play critical mechanical roles in neuronal function during cell proliferation, neuronal migration, and postmigrational development: the three major overlapping events of mammalian cerebral cortex development. A number of neuronally expressed tubulin genes are associated with a spectrum of disorders affecting cerebral cortex formation. Such "tubulinopathies" include lissencephaly/pachygyria, polymicrogyria-like malformations, and simplified gyral patterns, in addition to characteristic extracortical features, such as corpus callosal, basal ganglia, and cerebellar abnormalities. Epilepsy is a common finding in these related disorders. Here we describe two unrelated individuals with infantile-onset epilepsy and abnormalities of brain morphology, harboring de novo variants that affect adjacent amino acids in a beta-tubulin gene TUBB2A. Located in a highly conserved loop, we demonstrate impaired tubulin and microtubule function resulting from each variant in vitro and by using in silico predictive modeling. We propose that the affected functional loop directly associates with the alpha-tubulin-bound guanosine triphosphate (GTP) molecule, impairing the intradimer interface and correct formation of the alpha/beta-tubulin heterodimer. This study associates mutations in TUBB2A with the spectrum of "tubulinopathy" phenotypes. As a consequence, genetic variations affecting all beta-tubulin genes expressed at high levels in the brain (TUBB2B, TUBB3, TUBB, TUBB4A, and TUBB2A) have been linked with malformations of cortical development.
Collapse
Affiliation(s)
- Thomas D Cushion
- Neurology and Molecular Neuroscience Research Group, Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Alex R Paciorkowski
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA; Departments of Pediatrics and Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14641, USA; Center for Neural Development & Disease, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Daniela T Pilz
- Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK; Wales Epilepsy Research Network (WERN), College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Jonathan G L Mullins
- Neurology and Molecular Neuroscience Research Group, Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Laurie E Seltzer
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Robert W Marion
- The Children's Hospital at Montefiore, Bronx, NY 10467-2403, USA
| | - Emily Tuttle
- Center for Neural Development & Disease, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dalia Ghoneim
- Center for Neural Development & Disease, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Susan L Christian
- Center for Integrative Brain Research, Seattle Children's Hospital, Seattle, WA 98101, USA
| | - Seo-Kyung Chung
- Neurology and Molecular Neuroscience Research Group, Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK; Wales Epilepsy Research Network (WERN), College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Mark I Rees
- Neurology and Molecular Neuroscience Research Group, Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK; Wales Epilepsy Research Network (WERN), College of Medicine, Swansea University, Swansea SA2 8PP, UK.
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Hospital, Seattle, WA 98101, USA; Departments of Pediatrics and Neurology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
47
|
Zhang T, Zhao LL, Cao X, Qi LC, Wei GQ, Liu JY, Yan SJ, Liu JG, Li XQ. Bioinformatics analysis of time series gene expression in left ventricle (LV) with acute myocardial infarction (AMI). Gene 2014; 543:259-67. [PMID: 24704022 DOI: 10.1016/j.gene.2014.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/25/2014] [Accepted: 04/01/2014] [Indexed: 12/18/2022]
Abstract
This study is to investigate the key genes and their possible function in acute myocardial infarction (AMI). The data of GSE4648 downloaded from the Gene Expression Omnibus (GEO) database include 6 time points (15 min, 60 min, 4h, 12h, 24h and 48 h) of 12 left ventricle (LV) samples, 12 surviving LV free wall (FW) samples, 12 inter-ventricular septum (IVS) samples after AMI operation and corresponding sham-operated samples. The data of each sample were analyzed with Affy and Bioconductor packages, and differentially expressed genes (DEGs) were screened out using BETR package with false discovery rate (FDR)<0.01. Then, functional enrichment analysis for DEGs was conducted with Database for Annotation, Visualization and Integrated Discovery (DAVID). Totally 194 DEGs were identified in LV, and only the gene tubulin beta 2a (Tubb2a) and natriuretic peptide B (Nppb) were respectively up-regulated in surviving FW tissue and IVS tissue. The biological process response to wounding and inflammatory response were significantly enriched, as well as leukocyte transendothelial migration pathway. Besides, the expression pattern analysis showed the DEGs mostly up-regulated at 4h after AMI, and these genes were mainly associated with immunity. Additionally, in transcriptional regulatory network, early growth response 1 (Egr1), activating transcription factor 3 (Atf3), Atf4, Myc and Fos were considered as the key transcription factors related to immune response. The key transcription factors and potential target genes might provide new information for the development of AMI, and leukocyte transendothelial migration pathway might play a vital role in AMI.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Li-Li Zhao
- Department of Gastroenterology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xue Cao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Li-Chun Qi
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Guo-Qian Wei
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jun-Yan Liu
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Shu-Jun Yan
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jin-Gang Liu
- The Central Hospital of the Heilongjiang Prison Administrative Bureau, Harbin 150001, Heilongjiang Province, China
| | - Xue-Qi Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China.
| |
Collapse
|
48
|
Travis LB, Fossa SD, Sesso HD, Frisina RD, Herrmann DN, Beard CJ, Feldman DR, Pagliaro LC, Miller RC, Vaughn DJ, Einhorn LH, Cox NJ, Dolan ME. Chemotherapy-induced peripheral neurotoxicity and ototoxicity: new paradigms for translational genomics. J Natl Cancer Inst 2014; 106:dju044. [PMID: 24623533 PMCID: PMC4568989 DOI: 10.1093/jnci/dju044] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 01/07/2023] Open
Abstract
In view of advances in early detection and treatment, the 5-year relative survival rate for all cancer patients combined is now approximately 66%. As a result, there are more than 13.7 million cancer survivors in the United States, with this number increasing by 2% annually. For many patients, improvements in survival have been countered by therapy-associated adverse effects that may seriously impair long-term functional status, workplace productivity, and quality of life. Approximately 20% to 40% of cancer patients given neurotoxic chemotherapy develop chemotherapy-induced peripheral neurotoxicity (CIPN), which represents one of the most common and potentially permanent nonhematologic side effects of chemotherapy. Permanent bilateral hearing loss and/or tinnitus can result from several ototoxic therapies, including cisplatin- or carboplatin-based chemotherapy. CIPN and ototoxicity represent important challenges because of the lack of means for effective prevention, mitigation, or a priori identification of high-risk patients, and few studies have applied modern genomic approaches to understand underlying mechanisms/pathways. Translational genomics, including cell-based models, now offer opportunities to make inroads for the first time to develop preventive and interventional strategies for CIPN, ototoxicity, and other treatment-related complications. This commentary provides current perspective on a successful research strategy, with a focus on cisplatin, developed by an experienced, transdisciplinary group of researchers and clinicians, representing pharmacogenomics, statistical genetics, neurology, hearing science, medical oncology, epidemiology, and cancer survivorship. Principles outlined herein are applicable to the construction of research programs in translational genomics with strong clinical relevance and highlight unprecedented opportunities to understand, prevent, and treat long-term treatment-related morbidities.
Collapse
Affiliation(s)
- Lois B Travis
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL.
| | - Sophie D Fossa
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Howard D Sesso
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Robert D Frisina
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - David N Herrmann
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Clair J Beard
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Darren R Feldman
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Lance C Pagliaro
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Robert C Miller
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - David J Vaughn
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Lawrence H Einhorn
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Nancy J Cox
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - M Eileen Dolan
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| |
Collapse
|
49
|
Hertz DL. Germline pharmacogenetics of paclitaxel for cancer treatment. Pharmacogenomics 2014; 14:1065-84. [PMID: 23837481 DOI: 10.2217/pgs.13.90] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Paclitaxel is a highly effective chemotherapeutic agent used in a variety of solid tumors. Some paclitaxel-treated patients experience the intended therapeutic response with manageable side effects, while others have minimal response and/or severe toxicity. This variability in treatment outcome is partially determined by variability in drug exposure (pharmacokinetics) and by patient and tumor sensitivity (pharmacodynamics). Both pharmacokinetics and pharmacodynamics are dictated in part by common variants in the germline genome, known as SNPs. This article reviews the published literature on paclitaxel pharmacogenetics in cancer, focusing primarily on polymorphisms in genes relevant to paclitaxel pharmacokinetics and discusses preliminary work on pharmacodynamic genes and genome-wide association studies.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical, Social, & Administrative Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI, USA.
| |
Collapse
|
50
|
Jaeger S, Min J, Nigsch F, Camargo M, Hutz J, Cornett A, Cleaver S, Buckler A, Jenkins JL. Causal Network Models for Predicting Compound Targets and Driving Pathways in Cancer. ACTA ACUST UNITED AC 2014; 19:791-802. [PMID: 24518063 DOI: 10.1177/1087057114522690] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 01/14/2014] [Indexed: 02/06/2023]
Abstract
Gene-expression data are often used to infer pathways regulating transcriptional responses. For example, differentially expressed genes (DEGs) induced by compound treatment can help characterize hits from phenotypic screens, either by correlation with known drug signatures or by pathway enrichment. Pathway enrichment is, however, typically computed with DEGs rather than "upstream" nodes that are potentially causal of "downstream" changes. Here, we present graph-based models to predict causal targets from compound-microarray data. We test several approaches to traversing network topology, and show that a consensus minimum-rank score (SigNet) beat individual methods and could highly rank compound targets among all network nodes. In addition, larger, less canonical networks outperformed linear canonical interactions. Importantly, pathway enrichment using causal nodes rather than DEGs recovers relevant pathways more often. To further validate our approach, we used integrated data sets from the Cancer Genome Atlas to identify driving pathways in triple-negative breast cancer. Critical pathways were uncovered, including the epidermal growth factor receptor 2-phosphatidylinositide 3-kinase-AKT-MAPK growth pathway andATR-p53-BRCA DNA damage pathway, in addition to unexpected pathways, such as TGF-WNT cytoskeleton remodeling, IL12-induced interferon gamma production, and TNFR-IAP (inhibitor of apoptosis) apoptosis; the latter was validated by pooled small hairpin RNA profiling in cancer cells. Overall, our approach can bridge transcriptional profiles to compound targets and driving pathways in cancer.
Collapse
Affiliation(s)
- Savina Jaeger
- Co-first authors Oncology Translational Medicine, Novartis, Cambridge, MA, USA
| | - Junxia Min
- Co-first authors ONC Target Discovery, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - Florian Nigsch
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, Inc., Basel, Switzerland
| | - Miguel Camargo
- Center for Proteomic Chemistry, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - Janna Hutz
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA Pfizer, Cambridge, MA, USA
| | - Allen Cornett
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - Stephen Cleaver
- NIBR IT, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - Alan Buckler
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - Jeremy L Jenkins
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| |
Collapse
|