Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing

Diana Mandelker, Liying Zhang, Yelena Kemel, Zsofia K Stadler, Vijai Joseph, Ahmet Zehir, Nisha Pradhan, Angela Arnold, Michael F Walsh, Yirong Li, Anoop R Balakrishnan, Aijazuddin Syed, Meera Prasad, Khedoudja Nafa, Maria I Carlo, Karen A Cadoo, Meg Sheehan, Megan H Fleischut, Erin Salo-Mullen, Magan Trottier, Steven M Lipkin, Anne Lincoln, Semanti Mukherjee, Vignesh Ravichandran, Roy Cambria, Jesse Galle, Wassim Abida, Marcia E Arcila, Ryma Benayed, Ronak Shah, Kenneth Yu, Dean F Bajorin, Jonathan A Coleman, Steven D Leach, Maeve A Lowery, Julio Garcia-Aguilar, Philip W Kantoff, Charles L Sawyers, Maura N Dickler, Leonard Saltz, Robert J Motzer, Eileen M O'Reilly, Howard I Scher, Jose Baselga, David S Klimstra, David B Solit, David M Hyman, Michael F Berger, Marc Ladanyi, Mark E Robson, Kenneth Offit, Diana Mandelker, Liying Zhang, Yelena Kemel, Zsofia K Stadler, Vijai Joseph, Ahmet Zehir, Nisha Pradhan, Angela Arnold, Michael F Walsh, Yirong Li, Anoop R Balakrishnan, Aijazuddin Syed, Meera Prasad, Khedoudja Nafa, Maria I Carlo, Karen A Cadoo, Meg Sheehan, Megan H Fleischut, Erin Salo-Mullen, Magan Trottier, Steven M Lipkin, Anne Lincoln, Semanti Mukherjee, Vignesh Ravichandran, Roy Cambria, Jesse Galle, Wassim Abida, Marcia E Arcila, Ryma Benayed, Ronak Shah, Kenneth Yu, Dean F Bajorin, Jonathan A Coleman, Steven D Leach, Maeve A Lowery, Julio Garcia-Aguilar, Philip W Kantoff, Charles L Sawyers, Maura N Dickler, Leonard Saltz, Robert J Motzer, Eileen M O'Reilly, Howard I Scher, Jose Baselga, David S Klimstra, David B Solit, David M Hyman, Michael F Berger, Marc Ladanyi, Mark E Robson, Kenneth Offit

Abstract

Importance: Guidelines for cancer genetic testing based on family history may miss clinically actionable genetic changes with established implications for cancer screening or prevention.

Objective: To determine the proportion and potential clinical implications of inherited variants detected using simultaneous sequencing of the tumor and normal tissue ("tumor-normal sequencing") compared with genetic test results based on current guidelines.

Design, setting, and participants: From January 2014 until May 2016 at Memorial Sloan Kettering Cancer Center, 10 336 patients consented to tumor DNA sequencing. Since May 2015, 1040 of these patients with advanced cancer were referred by their oncologists for germline analysis of 76 cancer predisposition genes. Patients with clinically actionable inherited mutations whose genetic test results would not have been predicted by published decision rules were identified. Follow-up for potential clinical implications of mutation detection was through May 2017.

Exposure: Tumor and germline sequencing compared with the predicted yield of targeted germline sequencing based on clinical guidelines.

Main outcomes and measures: Proportion of clinically actionable germline mutations detected by universal tumor-normal sequencing that would not have been detected by guideline-directed testing.

Results: Of 1040 patients, the median age was 58 years (interquartile range, 50.5-66 years), 65.3% were male, and 81.3% had stage IV disease at the time of genomic analysis, with prostate, renal, pancreatic, breast, and colon cancer as the most common diagnoses. Of the 1040 patients, 182 (17.5%; 95% CI, 15.3%-19.9%) had clinically actionable mutations conferring cancer susceptibility, including 149 with moderate- to high-penetrance mutations; 101 patients tested (9.7%; 95% CI, 8.1%-11.7%) would not have had these mutations detected using clinical guidelines, including 65 with moderate- to high-penetrance mutations. Frequency of inherited mutations was related to case mix, stage, and founder mutations. Germline findings led to discussion or initiation of change to targeted therapy in 38 patients tested (3.7%) and predictive testing in the families of 13 individuals (1.3%), including 6 for whom genetic evaluation would not have been initiated by guideline-based testing.

Conclusions and relevance: In this referral population with selected advanced cancers, universal sequencing of a broad panel of cancer-related genes in paired germline and tumor DNA samples was associated with increased detection of individuals with potentially clinically significant heritable mutations over the predicted yield of targeted germline testing based on current clinical guidelines. Knowledge of these additional mutations can help guide therapeutic and preventive interventions, but whether all of these interventions would improve outcomes for patients with cancer or their family members requires further study.

Trial registration: clinicaltrials.gov Identifier: NCT01775072.

Conflict of interest statement

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Ms Kemel reported prior employment at Bioreference Laboratories. Dr Lowery reported serving on advisory boards for Agios Pharmaceuticals and Celgene. Dr Sawyers reporting serving on the board of directors for Novartis. Dr Dickler reported serving on advisory boards for Genentech/Roche, Novartis, Pfizer, Puma Biotech, AstraZeneca, and TapImmune. Dr Saltz reported receiving a grant from Taiho Pharmaceutical. Dr Scher reported serving as a consultant to Astellas, BIND Therapeutics, Clovis Oncology, Ferring Pharmaceuticals, Janssen Research and Development, Medivation, Merck, Roche, Sanofi Aventis, Takeda Millennium, and WIRB-Copernicus Group; receiving support to the Memorial Sloan Kettering Cancer Center from Medivation, Janssen, Illumina, and Innocrin Pharmaceuticals; and serving on the board of directors for Asterias Biotherapeutics. Dr Solit reported serving as a consultant to Pfizer and Loxo Oncology. Dr Hyman reported serving as a consultant to Chugai, CytomX, Boehringer Ingelheim, and Atara Biotherapeutics; and receiving grants from AstraZeneca, Puma Biotechnology, and Loxo Oncology. Dr Ladanyi reported receiving personal fees from Boehringer Ingelheim and AstraZeneca through the National Comprehensive Cancer Network; and a receiving a grant from Loxo Oncology. No other disclosures were reported.

Figures

Figure. Clinical Actionability, Concordance With Family History…
Figure. Clinical Actionability, Concordance With Family History and Phenotype, Penetrance, and Founder Mutations in 1040 Patients Undergoing Sequencing of Germline and Tumor DNA
Representation of the 1040 cases that carried clinically actionable pathogenic or presumed pathogenic variants, comprising 182 cases broken into subsets of no incremental cases (in which mutations would have been detected using genetic testing guidelines based on phenotype and family history) and incremental cases (in which mutations would not have been detected using guideline-based approaches). The incremental cases are categorized by high-, moderate-, or low-penetrance mutations. Numbers in parentheses indicate the numbers of patients in that category without Ashkenazi Jewish or European founder mutations.

References

    1. Schrader KA, Cheng DT, Joseph V, et al. Germline variants in targeted tumor sequencing using matched normal DNA. JAMA Oncol. 2016;2(1):104–111.
    1. Seifert BA, O’Daniel JM, Amin K, et al. Germline analysis from tumor-germline sequencing dyads to identify clinically actionable secondary findings. Clin Cancer Res. 2016;22(16):4087–4094.
    1. Jones S, Anagnostou V, Lytle K, et al. Personalized genomic analyses for cancer mutation discovery and interpretation. Sci Transl Med. 2015;7(283):283ra53.
    1. Parsons DW, Roy A, Yang Y, et al. Diagnostic yield of clinical tumor and germline whole-exome sequencing for children with solid tumors. JAMA Oncol. 2016;2(5):616–624.
    1. Mody RJ, Wu Y-M, Lonigro RJ, et al. Integrative clinical sequencing in the management of refractory or relapsed cancer in youth. JAMA. 2015;314(9):913–925.
    1. Meric-Bernstam F, Brusco L, Daniels M, et al. Incidental germline variants in 1000 advanced cancers on a prospective somatic genomic profiling protocol. Ann Oncol. 2016;27(5):795–800.
    1. Zhang J, Walsh MF, Wu G, et al. Germline mutations in predisposition genes in pediatric cancer. N Engl J Med. 2015;373(24):2336–2346.
    1. Hampel H, Bennett RL, Buchanan A, Pearlman R, Wiesner GL Guideline Development Group, American College of Medical Genetics and Genomics Professional Practice and Guidelines Committee and National Society of Genetic Counselors Practice Guidelines Committee. A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genet Med. 2015;17(1):70–87.
    1. National Comprehensive Cancer Network. [Accessed September 23, 2016];Genetic/Familial High-Risk Assessment: Breast and Ovarian, Version 1.2017. 2016 Sep 19; .
    1. National Comprehensive Cancer Network. [Accessed September 23, 2016];Genetic/Familial High-Risk Assessment: Colorectal, Version 2.2016. 2016 Sep 26; .
    1. Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn. 2015;17(3):251–264.
    1. Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23(6):703–713.
    1. Rahman N. Realizing the promise of cancer predisposition genes. Nature. 2014;505(7483):302–308.
    1. Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med. 2017;19(2):249–255.
    1. Cheng DT, Prasad M, Chekaluk Y, et al. Comprehensive detection of germline variants by MSK-IMPACT, a clinical diagnostic platform for solid tumor molecular oncology and concurrent cancer predisposition testing. BMC Med Genomics. 2017;10(1):33.
    1. Pritchard CC, Mateo J, Walsh MF, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med. 2016;375(5):443–453.
    1. Abida W, Armenia J, Gopalan A, et al. Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making. JCO Precis Oncol. 2017;(1):1–16. doi: 10.1200/PO.17.00029.
    1. Richards S, Aziz N, Bale S, et al. ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–424.
    1. Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016;44(16):e131.
    1. Niu B, Ye K, Zhang Q, et al. MSI sensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics. 2014;30(7):1015–1016.
    1. UpToDate. [Accessed October 19, 2016];Search. .
    1. American Society of Clinical Oncology. [Accessed October 19, 2016];Cancer Genetics Program. .
    1. Menko FH, van Steensel MAM, Giraud S, et al. European BHD Consortium. Birt-Hogg-Dubé syndrome: diagnosis and management. Lancet Oncol. 2009;10(12):1199–1206.
    1. Pilarski R, Cebulla CM, Massengill JB, et al. Expanding the clinical phenotype of hereditary BAP1 cancer predisposition syndrome, reporting three new cases. Genes Chromosomes Cancer. 2014;53(2):177–182.
    1. Thakker RV, Newey PJ, Walls GV, et al. Endocrine Society. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1) J Clin Endocrinol Metab. 2012;97(9):2990–3011.
    1. Exome Aggregation Consortium. [Accessed August 24, 2016];ExAC Browser Beta. .
    1. Lek M, Karczewski KJ, Minikel EV, et al. Exome Aggregation Consortium. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285–291.
    1. International Genome Sample Resource. [Accessed August 24, 2016];IGSR and the 1000 Genomes Project.
    1. National Center for Biotechnology Information. [Accessed August 24, 2016];dbSNP Short Genetic Variations.
    1. IBD Exomes Portal. [Accessed October 5, 2016];IBD Exomes browser.
    1. American Cancer Society. [Accessed December 19, 2016];Cancer facts and figures. 2016 .
    1. Forbes SA, Beare D, Gunasekaran P, et al. COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43(database issue):D805–D811.
    1. Wellcome Trust Sanger Institute Genome Research Ltd. [Accessed August 24, 2016];COSMIC: Catalogue of Somatic Mutations in Cancer. .
    1. Bombard Y, Robson M, Offit K. Revealing the incidentalome when targeting the tumor genome. JAMA. 2013;310(8):795–796.
    1. Wang LL, Gannavarapu A, Kozinetz CA, et al. Association between osteosarcoma and deleterious mutations in the RECQL4 gene in Rothmund-Thomson syndrome. J Natl Cancer Inst. 2003;95(9):669–674.
    1. Ng AJM, Walia MK, Smeets MF, et al. The DNA helicase RECQL4 is required for normal osteoblast expansion and osteosarcoma formation. PLoS Genet. 2015;11(4):e1005160.
    1. Wang LL, Plon SE. Rothmund-Thomson syndrome. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews. Seattle, WA: University of Washington; 1993.
    1. Offit K. The future of clinical cancer genomics. Semin Oncol. 2016;43(5):615–622.

Source: PubMed

3
Abonner