Oncotarget

Research Papers:

Rapid, ultra low coverage copy number profiling of cell-free DNA as a precision oncology screening strategy

Daniel H. Hovelson, Chia-Jen Liu, Yugang Wang, Qing Kang, James Henderson, Amy Gursky, Scott Brockman, Nithya Ramnath, John C. Krauss, Moshe Talpaz, Malathi Kandarpa, Rashmi Chugh, Missy Tuck, Kirk Herman, Catherine S. Grasso, Michael J. Quist, Felix Y. Feng, Christine Haakenson, John Langmore, Emmanuel Kamberov, Tim Tesmer, Hatim Husain, Robert J. Lonigro, Dan Robinson, David C. Smith, Ajjai S. Alva, Maha H. Hussain, Arul M. Chinnaiyan, Muneesh Tewari, Ryan E. Mills, Todd M. Morgan and Scott A. Tomlins _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:89848-89866. https://doi.org/10.18632/oncotarget.21163

Metrics: PDF 3651 views  |   HTML 7307 views  |   ?  


Abstract

Daniel H. Hovelson1,2, Chia-Jen Liu1,3, Yugang Wang4, Qing Kang5, James Henderson4, Amy Gursky4, Scott Brockman1, Nithya Ramnath5, John C. Krauss5, Moshe Talpaz5, Malathi Kandarpa5, Rashmi Chugh5, Missy Tuck5, Kirk Herman5, Catherine S. Grasso10,11, Michael J. Quist10,11, Felix Y. Feng12, Christine Haakenson13, John Langmore13, Emmanuel Kamberov13, Tim Tesmer13, Hatim Husain14, Robert J. Lonigro1,3, Dan Robinson1,3,8, David C. Smith5,8, Ajjai S. Alva5,8, Maha H. Hussain5,8,15, Arul M. Chinnaiyan1,3,8,10, Muneesh Tewari2,5,6,7,8,9, Ryan E. Mills2,7, Todd M. Morgan1,4,8,* and Scott A. Tomlins1,3,4,8,*

1Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA

2Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA

3Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA

4Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA

5Department of Internal Medicine (Hematology/Oncology), University of Michigan Medical School, Ann Arbor, MI, USA

6Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, USA

7Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA

8Department of Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA

9Department of Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, MI, USA

10Division of Hematology-Oncology, University of California, Los Angeles and the Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA

11The Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA

12Departments of Radiation Oncology, Urology, and Medicine, University of California at San Francisco, San Francisco, CA, USA

13Takara Bio USA, Ann Arbor, MI, USA

14Medical Oncology, University of California, San Diego Moore’s Cancer Center, San Diego, CA, USA

15Present address: Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA

*Co-senior authors

Correspondence to:

Scott A. Tomlins, email: [email protected]

Todd M. Morgan, email: [email protected]

Keywords: cell-free DNA, precision oncology, prostate cancer, whole genome sequencing, copy-number analysis

Received: August 29, 2017     Accepted: September 08, 2017     Published: September 22, 2017

ABSTRACT

Current cell-free DNA (cfDNA) next generation sequencing (NGS) precision oncology workflows are typically limited to targeted and/or disease-specific applications. In advanced cancer, disease burden and cfDNA tumor content are often elevated, yielding unique precision oncology opportunities. We sought to demonstrate the utility of a pan-cancer, rapid, inexpensive, whole genome NGS of cfDNA approach (PRINCe) as a precision oncology screening strategy via ultra-low coverage (~0.01x) tumor content determination through genome-wide copy number alteration (CNA) profiling. We applied PRINCe to a retrospective cohort of 124 cfDNA samples from 100 patients with advanced cancers, including 76 men with metastatic castration-resistant prostate cancer (mCRPC), enabling cfDNA tumor content approximation and actionable focal CNA detection, while facilitating concordance analyses between cfDNA and tissue-based NGS profiles and assessment of cfDNA alteration associations with mCRPC treatment outcomes. Therapeutically relevant focal CNAs were present in 42 (34%) cfDNA samples, including 36 of 93 (39%) mCRPC patient samples harboring AR amplification. PRINCe identified pre-treatment cfDNA CNA profiles facilitating disease monitoring. Combining PRINCe with routine targeted NGS of cfDNA enabled mutation and CNA assessment with coverages tuned to cfDNA tumor content. In mCRPC, genome-wide PRINCe cfDNA and matched tissue CNA profiles showed high concordance (median Pearson correlation = 0.87), and PRINCe detectable AR amplifications predicted reduced time on therapy, independent of therapy type (Kaplan-Meier log-rank test, chi-square = 24.9, p < 0.0001). Our screening approach enables robust, broadly applicable cfDNA-based precision oncology for patients with advanced cancer through scalable identification of therapeutically relevant CNAs and pre-/post-treatment genomic profiles, enabling cfDNA- or tissue-based precision oncology workflow optimization.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 21163