4.8 Article

Crowdsourced mapping of unexplored target space of kinase inhibitors

期刊

NATURE COMMUNICATIONS
卷 12, 期 1, 页码 -

出版社

NATURE PORTFOLIO
DOI: 10.1038/s41467-021-23165-1

关键词

-

资金

  1. Academy of Finland [310507, 313267, 326238]
  2. Cancer Research UK
  3. Brain Tumour Charity [REF: C42454/A28596]
  4. Helse SOr-Ost [2020026]
  5. National Institutes of Health [1U24DK116204-01, U54OD020353, U24CA224370, U24TR002278, U01CA239108]
  6. AbbVie
  7. Bayer Pharma AG
  8. Boehringer Ingelheim
  9. Canada Foundation for Innovation
  10. Eshelman Institute for Innovation
  11. Genome Canada
  12. Innovative Medicines Initiative [ULTRA-DD 115766]
  13. Wellcome Trust
  14. Janssen
  15. Merck Kga
  16. Merck Sharp Dohme
  17. Novartis Pharma AG
  18. Ontario Ministry of Economic Development and Innovation
  19. Pfizer
  20. Sao Paulo Research Foundation-FAPESP
  21. Takeda
  22. National Science Foundation (NSF) [CHE-1802789, CHE-2041108]
  23. Eshelman Institute for Innovation (EII) awards
  24. Molecular Sciences Software Institute (MolSSI) Software Fellowship
  25. NVIDIA Graduate Fellowship

向作者/读者索取更多资源

By benchmarking predictive algorithms on unpublished bioactivity data, it was found that ensemble models based on various approaches can improve prediction accuracy and accelerate experimental mapping of unexplored compound-kinase interactions.
Despite decades of intensive search for compounds that modulate the activity of particular protein targets, a large proportion of the human kinome remains as yet undrugged. Effective approaches are therefore required to map the massive space of unexplored compound-kinase interactions for novel and potent activities. Here, we carry out a crowdsourced benchmarking of predictive algorithms for kinase inhibitor potencies across multiple kinase families tested on unpublished bioactivity data. We find the top-performing predictions are based on various models, including kernel learning, gradient boosting and deep learning, and their ensemble leads to a predictive accuracy exceeding that of single-dose kinase activity assays. We design experiments based on the model predictions and identify unexpected activities even for under-studied kinases, thereby accelerating experimental mapping efforts. The open-source prediction algorithms together with the bioactivities between 95 compounds and 295 kinases provide a resource for benchmarking prediction algorithms and for extending the druggable kinome. The IDG-DREAM Challenge carried out crowdsourced benchmarking of predictive algorithms for kinase inhibitor activities on unpublished data. This study provides a resource to compare emerging algorithms and prioritize new kinase activities to accelerate drug discovery and repurposing efforts.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据