联合激酶组抑制状态可预测癌细胞系对激酶抑制剂联合疗法的敏感性。

Q2 Computer Science
Chinmaya U Joisa, Kevin A Chen, Samantha Beville, Timothy Stuhlmiller, Matthew E Berginski, Denis Okumu, Brian T Golitz, Michael P East, Gary L Johnson, Shawn M Gomez
{"title":"联合激酶组抑制状态可预测癌细胞系对激酶抑制剂联合疗法的敏感性。","authors":"Chinmaya U Joisa, Kevin A Chen, Samantha Beville, Timothy Stuhlmiller, Matthew E Berginski, Denis Okumu, Brian T Golitz, Michael P East, Gary L Johnson, Shawn M Gomez","doi":"","DOIUrl":null,"url":null,"abstract":"<p><p>Protein kinases are a primary focus in targeted therapy development for cancer, owing to their role as regulators in nearly all areas of cell life. Recent strategies targeting the kinome with combination therapies have shown promise, such as trametinib and dabrafenib in advanced melanoma, but empirical design for less characterized pathways remains a challenge. Computational combination screening is an attractive alternative, allowing in-silico filtering prior to experimental testing of drastically fewer leads, increasing efficiency and effectiveness of drug development pipelines. In this work, we generated combined kinome inhibition states of 40,000 kinase inhibitor combinations from kinobeads-based kinome profiling across 64 doses. We then integrated these with transcriptomics from CCLE to build machine learning models with elastic-net feature selection to predict cell line sensitivity across nine cancer types, with accuracy R2 ∼ 0.75-0.9. We then validated the model by using a PDX-derived TNBC cell line and saw good global accuracy (R2 ∼ 0.7) as well as high accuracy in predicting synergy using four popular metrics (R2 ∼ 0.9). Additionally, the model was able to predict a highly synergistic combination of trametinib and omipalisib for TNBC treatment, which incidentally was recently in phase I clinical trials. Our choice of tree-based models for greater interpretability allowed interrogation of highly predictive kinases in each cancer type, such as the MAPK, CDK, and STK kinases. Overall, these results suggest that kinome inhibition states of kinase inhibitor combinations are strongly predictive of cell line responses and have great potential for integration into computational drug screening pipelines. This approach may facilitate the identification of effective kinase inhibitor combinations and accelerate the development of novel cancer therapies, ultimately improving patient outcomes.</p>","PeriodicalId":34954,"journal":{"name":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413988/pdf/","citationCount":"0","resultStr":"{\"title\":\"Combined kinome inhibition states are predictive of cancer cell line sensitivity to kinase inhibitor combination therapies.\",\"authors\":\"Chinmaya U Joisa, Kevin A Chen, Samantha Beville, Timothy Stuhlmiller, Matthew E Berginski, Denis Okumu, Brian T Golitz, Michael P East, Gary L Johnson, Shawn M Gomez\",\"doi\":\"\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Protein kinases are a primary focus in targeted therapy development for cancer, owing to their role as regulators in nearly all areas of cell life. Recent strategies targeting the kinome with combination therapies have shown promise, such as trametinib and dabrafenib in advanced melanoma, but empirical design for less characterized pathways remains a challenge. Computational combination screening is an attractive alternative, allowing in-silico filtering prior to experimental testing of drastically fewer leads, increasing efficiency and effectiveness of drug development pipelines. In this work, we generated combined kinome inhibition states of 40,000 kinase inhibitor combinations from kinobeads-based kinome profiling across 64 doses. We then integrated these with transcriptomics from CCLE to build machine learning models with elastic-net feature selection to predict cell line sensitivity across nine cancer types, with accuracy R2 ∼ 0.75-0.9. We then validated the model by using a PDX-derived TNBC cell line and saw good global accuracy (R2 ∼ 0.7) as well as high accuracy in predicting synergy using four popular metrics (R2 ∼ 0.9). Additionally, the model was able to predict a highly synergistic combination of trametinib and omipalisib for TNBC treatment, which incidentally was recently in phase I clinical trials. Our choice of tree-based models for greater interpretability allowed interrogation of highly predictive kinases in each cancer type, such as the MAPK, CDK, and STK kinases. Overall, these results suggest that kinome inhibition states of kinase inhibitor combinations are strongly predictive of cell line responses and have great potential for integration into computational drug screening pipelines. This approach may facilitate the identification of effective kinase inhibitor combinations and accelerate the development of novel cancer therapies, ultimately improving patient outcomes.</p>\",\"PeriodicalId\":34954,\"journal\":{\"name\":\"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-01-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413988/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"Computer Science\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing","FirstCategoryId":"1085","ListUrlMain":"","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"Computer Science","Score":null,"Total":0}
引用次数: 0

摘要

蛋白激酶是癌症靶向疗法开发的主要焦点,因为它们在细胞生命的几乎所有领域都发挥着调节作用。最近以激酶组为靶点的联合疗法策略已初见成效,如用于晚期黑色素瘤的曲美替尼和达拉非尼,但针对特征较少的通路进行经验性设计仍是一项挑战。计算组合筛选是一种有吸引力的替代方法,它可以在实验测试之前对数量大幅减少的线索进行体内筛选,从而提高药物开发流水线的效率和有效性。在这项工作中,我们通过基于激酶标靶的激酶组图谱分析,生成了 40,000 种激酶抑制剂组合在 64 种剂量下的综合激酶组抑制状态。然后,我们将其与 CCLE 的转录组学整合,建立了具有弹性网特征选择的机器学习模型,以预测九种癌症类型的细胞系敏感性,准确率 R2 ∼ 0.75-0.9。然后,我们使用源自 TNBC 细胞系的 PDX 验证了该模型,结果显示该模型具有良好的全局准确性(R2 ∼ 0.7),而且使用四种常用指标预测协同作用的准确性也很高(R2 ∼ 0.9)。此外,该模型还能预测曲美替尼和奥米帕利在 TNBC 治疗中的高度协同组合,而这一组合最近刚刚进入 I 期临床试验。我们选择基于树状结构的模型以提高可解释性,这样就能对每种癌症类型中的高预测性激酶进行分析,如 MAPK、CDK 和 STK 激酶。总之,这些结果表明,激酶抑制剂组合的激酶组抑制状态对细胞系反应具有很强的预测性,并具有整合到计算药物筛选管道的巨大潜力。这种方法可以促进有效激酶抑制剂组合的鉴定,加快新型癌症疗法的开发,最终改善患者的预后。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Combined kinome inhibition states are predictive of cancer cell line sensitivity to kinase inhibitor combination therapies.

Protein kinases are a primary focus in targeted therapy development for cancer, owing to their role as regulators in nearly all areas of cell life. Recent strategies targeting the kinome with combination therapies have shown promise, such as trametinib and dabrafenib in advanced melanoma, but empirical design for less characterized pathways remains a challenge. Computational combination screening is an attractive alternative, allowing in-silico filtering prior to experimental testing of drastically fewer leads, increasing efficiency and effectiveness of drug development pipelines. In this work, we generated combined kinome inhibition states of 40,000 kinase inhibitor combinations from kinobeads-based kinome profiling across 64 doses. We then integrated these with transcriptomics from CCLE to build machine learning models with elastic-net feature selection to predict cell line sensitivity across nine cancer types, with accuracy R2 ∼ 0.75-0.9. We then validated the model by using a PDX-derived TNBC cell line and saw good global accuracy (R2 ∼ 0.7) as well as high accuracy in predicting synergy using four popular metrics (R2 ∼ 0.9). Additionally, the model was able to predict a highly synergistic combination of trametinib and omipalisib for TNBC treatment, which incidentally was recently in phase I clinical trials. Our choice of tree-based models for greater interpretability allowed interrogation of highly predictive kinases in each cancer type, such as the MAPK, CDK, and STK kinases. Overall, these results suggest that kinome inhibition states of kinase inhibitor combinations are strongly predictive of cell line responses and have great potential for integration into computational drug screening pipelines. This approach may facilitate the identification of effective kinase inhibitor combinations and accelerate the development of novel cancer therapies, ultimately improving patient outcomes.

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
CiteScore
4.50
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