Deep-Learning Model for Tumor-Type Prediction Using Targeted Clinical Genomic Sequencing Data

Author:

Darmofal Madison12ORCID,Suman Shalabh3ORCID,Atwal Gurnit456ORCID,Toomey Michael12ORCID,Chen Jie-Fu3ORCID,Chang Jason C.3ORCID,Vakiani Efsevia3ORCID,Varghese Anna M.7ORCID,Balakrishnan Rema Anoop3ORCID,Syed Aijazuddin3ORCID,Schultz Nikolaus8910ORCID,Berger Michael F.389ORCID,Morris Quaid1ORCID

Affiliation:

1. 1Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York.

2. 2Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, New York.

3. 3Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.

4. 4Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.

5. 5Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.

6. 6Vector Institute, Toronto, Ontario, Canada.

7. 7Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.

8. 8Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.

9. 9Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.

10. 10Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York.

Abstract

Abstract Tumor type guides clinical treatment decisions in cancer, but histology-based diagnosis remains challenging. Genomic alterations are highly diagnostic of tumor type, and tumor-type classifiers trained on genomic features have been explored, but the most accurate methods are not clinically feasible, relying on features derived from whole-genome sequencing (WGS), or predicting across limited cancer types. We use genomic features from a data set of 39,787 solid tumors sequenced using a clinically targeted cancer gene panel to develop Genome-Derived-Diagnosis Ensemble (GDD-ENS): a hyperparameter ensemble for classifying tumor type using deep neural networks. GDD-ENS achieves 93% accuracy for high-confidence predictions across 38 cancer types, rivaling the performance of WGS-based methods. GDD-ENS can also guide diagnoses of rare type and cancers of unknown primary and incorporate patient-specific clinical information for improved predictions. Overall, integrating GDD-ENS into prospective clinical sequencing workflows could provide clinically relevant tumor-type predictions to guide treatment decisions in real time. Significance: We describe a highly accurate tumor-type prediction model, designed specifically for clinical implementation. Our model relies only on widely used cancer gene panel sequencing data, predicts across 38 distinct cancer types, and supports integration of patient-specific nongenomic information for enhanced decision support in challenging diagnostic situations. See related commentary by Garg, p. 906. This article is featured in Selected Articles from This Issue, p. 897

Funder

National Cancer Institute

Publisher

American Association for Cancer Research (AACR)

Cited by 2 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3