Diagnostic Yield of Clinical Tumor and Germline Whole-Exome Sequencing for Children With Solid Tumors.

Importance Whole-exome sequencing (WES) has the potential to reveal tumor and germline mutations of clinical relevance, but the diagnostic yield for pediatric patients with solid tumors is unknown. Objective To characterize the diagnostic yield of combined tumor and germline WES for children with solid tumors. Design Unselected children with newly diagnosed and previously untreated central nervous system (CNS) and non-CNS solid tumors were prospectively enrolled in the BASIC3 study at a large academic children's hospital during a 23-month period from August 2012 through June 2014. Blood and tumor samples underwent WES in a certified clinical laboratory with genetic results categorized on the basis of perceived clinical relevance and entered in the electronic health record. Main Outcomes and Measures Clinical categorization of somatic mutations; frequencies of deleterious germline mutations related to patient phenotype and incidental medically-actionable mutations. Results Of the first 150 participants (80 boys and 70 girls, mean age, 7.4 years), tumor samples adequate for WES were available from 121 patients (81%). Somatic mutations of established clinical utility (category I) were reported in 4 (3%) of 121 patients, with mutations of potential utility (category II) detected in an additional 29 (24%) of 121 patients. CTNNB1 was the gene most frequently mutated, with recurrent mutations in KIT, TSC2, and MAPK pathway genes (BRAF, KRAS, and NRAS) also identified. Mutations in consensus cancer genes (category III) were found in an additional 24 (20%) of 121 tumors. Fewer than half of somatic mutations identified were in genes known to be recurrently mutated in the tumor type tested. Diagnostic germline findings related to patient phenotype were discovered in 15 (10%) of 150 cases: 13 pathogenic or likely pathogenic dominant mutations in adult and pediatric cancer susceptibility genes (including 2 each in TP53, VHL, and BRCA1), 1 recessive liver disorder with hepatocellular carcinoma (TJP2), and 1 renal diagnosis (CLCN5). Incidental findings were reported in 8 (5%) of 150 patients. Most patients harbored germline uncertain variants in cancer genes (98%), pharmacogenetic variants (89%), and recessive carrier mutations (85%). Conclusions and Relevance Tumor and germline WES revealed mutations in a broad spectrum of genes previously implicated in both adult and pediatric cancers. Combined reporting of tumor and germline WES identified diagnostic and/or potentially actionable findings in nearly 40% of newly diagnosed pediatric patients with solid tumors.

[1]  H. Witt,et al.  Pediatric Brain Tumors: Innovative Genomic Information Is Transforming the Diagnostic and Clinical Landscape. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[2]  Theresa Zhang,et al.  Personalized genomic analyses for cancer mutation discovery and interpretation , 2015, Science Translational Medicine.

[3]  A. Redig,et al.  Basket trials and the evolution of clinical trial design in an era of genomic medicine. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  Magalie S Leduc,et al.  Molecular findings among patients referred for clinical whole-exome sequencing. , 2014, JAMA.

[5]  Mingming Jia,et al.  COSMIC: exploring the world's knowledge of somatic mutations in human cancer , 2014, Nucleic Acids Res..

[6]  Adam Kiezun,et al.  Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma , 2014, Proceedings of the National Academy of Sciences.

[7]  R. Gibbs,et al.  Obtaining informed consent for clinical tumor and germline exome sequencing of newly diagnosed childhood cancer patients , 2014, Genome Medicine.

[8]  Leslie G Biesecker,et al.  Diagnostic clinical genome and exome sequencing. , 2014, The New England journal of medicine.

[9]  Yuya Kobayashi,et al.  Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[10]  R. Gibbs,et al.  Launching genomics into the cloud: deployment of Mercury, a next generation sequence analysis pipeline , 2014, BMC Bioinformatics.

[11]  Benjamin J. Raphael,et al.  Integrated Analysis of Germline and Somatic Variants in Ovarian Cancer , 2014, Nature Communications.

[12]  Magalie S Leduc,et al.  Clinical whole-exome sequencing for the diagnosis of mendelian disorders. , 2013, The New England journal of medicine.

[13]  Emily H Turner,et al.  Actionable, pathogenic incidental findings in 1,000 participants' exomes. , 2013, American journal of human genetics.

[14]  Manash S. Chatterjee,et al.  The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. , 2013, The Lancet. Oncology.

[15]  Marc S. Williams,et al.  ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing , 2013, Genetics in Medicine.

[16]  Levi A Garraway,et al.  Genomics-driven oncology: framework for an emerging paradigm. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[17]  K. Kinzler,et al.  Cancer Genome Landscapes , 2013, Science.

[18]  Steven J. M. Jones,et al.  The genetic landscape of high-risk neuroblastoma , 2013, Nature Genetics.

[19]  Giovanni Parmigiani,et al.  Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma , 2012, Nature Genetics.

[20]  C. Sander,et al.  Genome Sequencing Identifies a Basis for Everolimus Sensitivity , 2012, Science.

[21]  Jill P. Mesirov,et al.  MEDULLOBLASTOMA EXOME SEQUENCING UNCOVERS SUBTYPE-SPECIFIC SOMATIC MUTATIONS , 2012, Nature.

[22]  Elaine R. Mardis,et al.  Novel mutations target distinct subgroups of medulloblastoma , 2012, Nature.

[23]  J. Opitz,et al.  Germline mutations in DIS3L2 cause the Perlman syndrome of overgrowth and Wilms tumor susceptibility , 2012, Nature Genetics.

[24]  R. Gibbs,et al.  Targeted enrichment beyond the consensus coding DNA sequence exome reveals exons with higher variant densities , 2011, Genome Biology.

[25]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[26]  A. Byars,et al.  Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. , 2010, The New England journal of medicine.

[27]  R. Siebert,et al.  Germline nonsense mutation and somatic inactivation of SMARCA4/BRG1 in a family with rhabdoid tumor predisposition syndrome. , 2010, American journal of human genetics.

[28]  Nazneen Rahman,et al.  Common variations in BARD1 influence susceptibility to high-risk neuroblastoma , 2009, Nature Genetics.

[29]  K. Nathanson,et al.  Childhood cancer in families with and without BRCA1 or BRCA2 mutations ascertained at a high-risk breast cancer clinic , 2006, Cancer biology & therapy.

[30]  Adopted on March American Society of Clinical Oncology policy statement update: genetic testing for cancer susceptibility. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.