定量研究体细胞突变对癌症的影响。

Oncoscience Pub Date : 2020-10-27 eCollection Date: 2020-11-01 DOI:10.18632/oncoscience.521
Jeffrey P Townsend
{"title":"定量研究体细胞突变对癌症的影响。","authors":"Jeffrey P Townsend","doi":"10.18632/oncoscience.521","DOIUrl":null,"url":null,"abstract":"Numerous powerful bioinformatic analyses of cancer tumor sequencing have applied sophisticated mutation calling, determining the key cancer-causing variants and quantifying their prevalence. The calculations of prevalence of a mutation across tumors and the determination of the statistical significance of whether it is a driver are the “shoulders” that have enabled the build-up to the most useful metrics about cancer variants—metrics which quantify the effect of the variant on replication and survival of the cancer lineage. Ostrow et al [1] effectively and comprehensively applied ratios of non-synonymous change to synonymous change to quantify natural selection in the somatic evolution of cancer, an approach that has been followed by others in different ways and contexts since then [2–4]. Martincorena et al [2] performed a cogent gene-wide analysis using mutation signatures c.f. [5] on the larger data sets available three years later. More recently, it was revealed that previous studies have reported variant prevalence and P value, but have not reported cancer effect sizes, which quantify the effect of natural selection on somatic mutations within cancer cell lineages. Deconvolving the baseline variant mutation rates enables estimation of the selection intensity of individual mutations [6, 7]—a quantification that should be directly interpreted as a cancer lineage effect size that should be used in decision-making. This measure of the effect of specific somatic mutations on cancer cell proliferation and survival should be widely appreciated as a primary consideration of precisionmedicine tumor boards, which are in operation at hospitals around the world. Effect sizes of somatic mutations should also be a key consideration in the initiation and design of precision medicine clinical trials: the number of trials has been increasing so rapidly that some have argued that demand is vastly outpacing the supply of enrollable patients [8]. Effect sizes should guide target selection in pharmacological development, an approximately three billion dollar industry [9]. And gene-specific site-specific effect sizes should guide basic research prioritization toward those important components of molecular and cell biology that have long-term potential to lead to therapies and cures for cancer. There appear to be increasing numbers of drivers in each cancer as we examine larger and larger datasets [10], and each driver has its own quantitative effect on cancer [11]. Decoupling the contributions of mutation and cancer lineage selection to the frequency of somatic variants among tumors is critical to understanding— and predicting—the therapeutic potential of different interventions [12]. Importantly, antagonistic and synergistic epistasis among mutations also impacts the potential therapeutic benefit of targeted drug development [13]. Active use of these quantitative approaches are essential to furthering basic research on cancer, informing clinical practice, and providing rigorous guidance regarding investment in targeted drug development. By integrating evolutionary principles and detailed mechanistic knowledge into those approaches, we can maximize our ability to apply and develop targeted therapies that are likely to yield substantial clinical benefit.","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2020-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7781488/pdf/","citationCount":"0","resultStr":"{\"title\":\"Getting quantitative on the effects of somatic mutation on cancer.\",\"authors\":\"Jeffrey P Townsend\",\"doi\":\"10.18632/oncoscience.521\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Numerous powerful bioinformatic analyses of cancer tumor sequencing have applied sophisticated mutation calling, determining the key cancer-causing variants and quantifying their prevalence. The calculations of prevalence of a mutation across tumors and the determination of the statistical significance of whether it is a driver are the “shoulders” that have enabled the build-up to the most useful metrics about cancer variants—metrics which quantify the effect of the variant on replication and survival of the cancer lineage. Ostrow et al [1] effectively and comprehensively applied ratios of non-synonymous change to synonymous change to quantify natural selection in the somatic evolution of cancer, an approach that has been followed by others in different ways and contexts since then [2–4]. Martincorena et al [2] performed a cogent gene-wide analysis using mutation signatures c.f. [5] on the larger data sets available three years later. More recently, it was revealed that previous studies have reported variant prevalence and P value, but have not reported cancer effect sizes, which quantify the effect of natural selection on somatic mutations within cancer cell lineages. Deconvolving the baseline variant mutation rates enables estimation of the selection intensity of individual mutations [6, 7]—a quantification that should be directly interpreted as a cancer lineage effect size that should be used in decision-making. This measure of the effect of specific somatic mutations on cancer cell proliferation and survival should be widely appreciated as a primary consideration of precisionmedicine tumor boards, which are in operation at hospitals around the world. Effect sizes of somatic mutations should also be a key consideration in the initiation and design of precision medicine clinical trials: the number of trials has been increasing so rapidly that some have argued that demand is vastly outpacing the supply of enrollable patients [8]. Effect sizes should guide target selection in pharmacological development, an approximately three billion dollar industry [9]. And gene-specific site-specific effect sizes should guide basic research prioritization toward those important components of molecular and cell biology that have long-term potential to lead to therapies and cures for cancer. There appear to be increasing numbers of drivers in each cancer as we examine larger and larger datasets [10], and each driver has its own quantitative effect on cancer [11]. Decoupling the contributions of mutation and cancer lineage selection to the frequency of somatic variants among tumors is critical to understanding— and predicting—the therapeutic potential of different interventions [12]. Importantly, antagonistic and synergistic epistasis among mutations also impacts the potential therapeutic benefit of targeted drug development [13]. Active use of these quantitative approaches are essential to furthering basic research on cancer, informing clinical practice, and providing rigorous guidance regarding investment in targeted drug development. By integrating evolutionary principles and detailed mechanistic knowledge into those approaches, we can maximize our ability to apply and develop targeted therapies that are likely to yield substantial clinical benefit.\",\"PeriodicalId\":19508,\"journal\":{\"name\":\"Oncoscience\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2020-10-27\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7781488/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Oncoscience\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.18632/oncoscience.521\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2020/11/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Oncoscience","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.18632/oncoscience.521","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2020/11/1 0:00:00","PubModel":"eCollection","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

本文章由计算机程序翻译,如有差异,请以英文原文为准。
Getting quantitative on the effects of somatic mutation on cancer.
Numerous powerful bioinformatic analyses of cancer tumor sequencing have applied sophisticated mutation calling, determining the key cancer-causing variants and quantifying their prevalence. The calculations of prevalence of a mutation across tumors and the determination of the statistical significance of whether it is a driver are the “shoulders” that have enabled the build-up to the most useful metrics about cancer variants—metrics which quantify the effect of the variant on replication and survival of the cancer lineage. Ostrow et al [1] effectively and comprehensively applied ratios of non-synonymous change to synonymous change to quantify natural selection in the somatic evolution of cancer, an approach that has been followed by others in different ways and contexts since then [2–4]. Martincorena et al [2] performed a cogent gene-wide analysis using mutation signatures c.f. [5] on the larger data sets available three years later. More recently, it was revealed that previous studies have reported variant prevalence and P value, but have not reported cancer effect sizes, which quantify the effect of natural selection on somatic mutations within cancer cell lineages. Deconvolving the baseline variant mutation rates enables estimation of the selection intensity of individual mutations [6, 7]—a quantification that should be directly interpreted as a cancer lineage effect size that should be used in decision-making. This measure of the effect of specific somatic mutations on cancer cell proliferation and survival should be widely appreciated as a primary consideration of precisionmedicine tumor boards, which are in operation at hospitals around the world. Effect sizes of somatic mutations should also be a key consideration in the initiation and design of precision medicine clinical trials: the number of trials has been increasing so rapidly that some have argued that demand is vastly outpacing the supply of enrollable patients [8]. Effect sizes should guide target selection in pharmacological development, an approximately three billion dollar industry [9]. And gene-specific site-specific effect sizes should guide basic research prioritization toward those important components of molecular and cell biology that have long-term potential to lead to therapies and cures for cancer. There appear to be increasing numbers of drivers in each cancer as we examine larger and larger datasets [10], and each driver has its own quantitative effect on cancer [11]. Decoupling the contributions of mutation and cancer lineage selection to the frequency of somatic variants among tumors is critical to understanding— and predicting—the therapeutic potential of different interventions [12]. Importantly, antagonistic and synergistic epistasis among mutations also impacts the potential therapeutic benefit of targeted drug development [13]. Active use of these quantitative approaches are essential to furthering basic research on cancer, informing clinical practice, and providing rigorous guidance regarding investment in targeted drug development. By integrating evolutionary principles and detailed mechanistic knowledge into those approaches, we can maximize our ability to apply and develop targeted therapies that are likely to yield substantial clinical benefit.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信