Large-scale cancer sequencing efforts provide a unique opportunity for the discovery of germline and somatic driver alterations influencing cancer susceptibility, initiation, progression, and clinical response. Detecting such alterations is fundamentally and technically challenging for several reasons including: 1) the combinatorially enormous number of ways that a genome can be altered, 2) the presence of various sized repeats, highly homologous gene families, and other contextual influences on alignment and detection accuracy, 3) systematic errors inherent in current sequencing technologies and tumor preservation techniques, and 4) intratumoral and intertumoral heterogeneity including clonality, purity, and lymphocyte infiltration. As a result, the full complement of driver events for the typical tumor still defies identification and, in many cases, no drivers can be found. Our recent work has also demonstrated that some types of indels/SVs such as complex indels, ITD/PTD (internal/partial tandem duplications), and homopolymer indels are often missed by existing approaches. Beyond detection challenges, functional interpretation of the impact of genomic alterations requires strategies that integrate WGS/exome, RNA-seq, and protein data to reveal translational, splicing, and protein structural effects. In addition, cooperative dynamics between germline and somatic alterations are usually missed, as these events have been analyzed independently. As cancer sequencing projects expand to include well-curated clinical phenotypes, methods necessary to understand the pathogenicity and druggability of driver alterations that underlie phenotypes such as drug resistance or exceptional responders are also urgently needed. To fully harness the power of large-scale cancer genomics and to facilitate advances in personalized medicine, our group proposes to focus on two core competencies, coding and non-coding mutations, outlined in the RFA. In collaboration with other GDACs, GDC, and AWGs, we will extend computational approaches that we have successfully established and applied for TCGA and ICGC projects to detect and functionally and clinically interpret germline and somatic drivers using sequencing data from GCC along with curated clinical data.

Public Health Relevance

Understanding the whole spectrum of inherited and acquired genetic changes in cancer will lead to effective diagnosis and treatment strategies for each patient's cancer. Development of advanced bioinformatics approaches will be a prerequisite for successful diagnosis, treatment, and management of cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Resource-Related Research Projects--Cooperative Agreements (U24)
Project #
1U24CA211006-01
Application #
9211152
Study Section
Special Emphasis Panel (ZCA1-SRB-L (A1))
Program Officer
Yang, Liming
Project Start
2016-09-14
Project End
2021-08-31
Budget Start
2016-09-14
Budget End
2017-08-31
Support Year
1
Fiscal Year
2016
Total Cost
$391,021
Indirect Cost
$66,919
Name
Washington University
Department
Genetics
Type
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Bailey, Matthew H; Tokheim, Collin; Porta-Pardo, Eduard et al. (2018) Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 173:371-385.e18
Sengupta, Sohini; Sun, Sam Q; Huang, Kuan-Lin et al. (2018) Integrative omics analyses broaden treatment targets in human cancer. Genome Med 10:60
Huang, Kuan-Lin; Mashl, R Jay; Wu, Yige et al. (2018) Pathogenic Germline Variants in 10,389 Adult Cancers. Cell 173:355-370.e14
Ding, Li; Bailey, Matthew H; Porta-Pardo, Eduard et al. (2018) Perspective on Oncogenic Processes at the End of the Beginning of Cancer Genomics. Cell 173:305-320.e10
Jayasinghe, Reyka G; Cao, Song; Gao, Qingsong et al. (2018) Systematic Analysis of Splice-Site-Creating Mutations in Cancer. Cell Rep 23:270-281.e3
Devarakonda, Siddhartha; Govindan, Ramaswamy (2018) Targeting Resistance to Targeted Therapies: Combating a Resilient Foe. Clin Cancer Res 24:6112-6114
Ellrott, Kyle; Bailey, Matthew H; Saksena, Gordon et al. (2018) Scalable Open Science Approach for Mutation Calling of Tumor Exomes Using Multiple Genomic Pipelines. Cell Syst 6:271-281.e7
Gao, Qingsong; Liang, Wen-Wei; Foltz, Steven M et al. (2018) Driver Fusions and Their Implications in the Development and Treatment of Human Cancers. Cell Rep 23:227-238.e3
Ding, Li; Chen, Feng (2017) Precancerous Clonal Expansion: A New Therapeutic Target? J Clin Oncol 35:1503-1505
Huang, Kuan-Lin; Li, Shunqiang; Mertins, Philipp et al. (2017) Proteogenomic integration reveals therapeutic targets in breast cancer xenografts. Nat Commun 8:14864

Showing the most recent 10 out of 17 publications