Clinical interpretation and implications of whole-genome sequencing.

IMPORTANCE Whole-genome sequencing (WGS) is increasingly applied in clinical medicine and is expected to uncover clinically significant findings regardless of sequencing indication. OBJECTIVES To examine coverage and concordance of clinically relevant genetic variation provided by WGS technologies; to quantitate inherited disease risk and pharmacogenomic findings in WGS data and resources required for their discovery and interpretation; and to evaluate clinical action prompted by WGS findings. DESIGN, SETTING, AND PARTICIPANTS An exploratory study of 12 adult participants recruited at Stanford University Medical Center who underwent WGS between November 2011 and March 2012. A multidisciplinary team reviewed all potentially reportable genetic findings. Five physicians proposed initial clinical follow-up based on the genetic findings. MAIN OUTCOMES AND MEASURES Genome coverage and sequencing platform concordance in different categories of genetic disease risk, person-hours spent curating candidate disease-risk variants, interpretation agreement between trained curators and disease genetics databases, burden of inherited disease risk and pharmacogenomic findings, and burden and interrater agreement of proposed clinical follow-up. RESULTS Depending on sequencing platform, 10% to 19% of inherited disease genes were not covered to accepted standards for single nucleotide variant discovery. Genotype concordance was high for previously described single nucleotide genetic variants (99%-100%) but low for small insertion/deletion variants (53%-59%). Curation of 90 to 127 genetic variants in each participant required a median of 54 minutes (range, 5-223 minutes) per genetic variant, resulted in moderate classification agreement between professionals (Gross κ, 0.52; 95% CI, 0.40-0.64), and reclassified 69% of genetic variants cataloged as disease causing in mutation databases to variants of uncertain or lesser significance. Two to 6 personal disease-risk findings were discovered in each participant, including 1 frameshift deletion in the BRCA1 gene implicated in hereditary breast and ovarian cancer. Physician review of sequencing findings prompted consideration of a median of 1 to 3 initial diagnostic tests and referrals per participant, with fair interrater agreement about the suitability of WGS findings for clinical follow-up (Fleiss κ, 0.24; P < 001). CONCLUSIONS AND RELEVANCE In this exploratory study of 12 volunteer adults, the use of WGS was associated with incomplete coverage of inherited disease genes, low reproducibility of detection of genetic variation with the highest potential clinical effects, and uncertainty about clinically reportable findings. In certain cases, WGS will identify clinically actionable genetic variants warranting early medical intervention. These issues should be considered when determining the role of WGS in clinical medicine.

[1]  David P Bick,et al.  Making a definitive diagnosis: Successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease , 2011, Genetics in Medicine.

[2]  J. Fleiss Measuring nominal scale agreement among many raters. , 1971 .

[3]  Chong Shen,et al.  Genome-wide association study in Han Chinese identifies four new susceptibility loci for coronary artery disease , 2012, Nature Genetics.

[4]  Euan A Ashley,et al.  A public resource facilitating clinical use of genomes , 2012, Proceedings of the National Academy of Sciences.

[5]  T E Klein,et al.  Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for Codeine Therapy in the Context of Cytochrome P450 2D6 (CYP2D6) Genotype , 2012, Clinical pharmacology and therapeutics.

[6]  Jonathan C. Cohen,et al.  Exome sequencing, ANGPTL3 mutations, and familial combined hypolipidemia. , 2010, The New England journal of medicine.

[7]  Patrizia Sola,et al.  Exome Sequencing Reveals VCP Mutations as a Cause of Familial ALS , 2011, Neuron.

[8]  R. Altman,et al.  Pharmacogenomics Knowledge for Personalized Medicine , 2012, Clinical pharmacology and therapeutics.

[9]  Nancy F. Hansen,et al.  Accurate Whole Human Genome Sequencing using Reversible Terminator Chemistry , 2008, Nature.

[10]  D. MacArthur,et al.  Loss-of-function variants in the genomes of healthy humans. , 2010, Human molecular genetics.

[11]  L. Esserman,et al.  Genetic/familial high-risk assessment , 2010 .

[12]  Larry N. Singh,et al.  Secondary variants in individuals undergoing exome sequencing: screening of 572 individuals identifies high-penetrance mutations in cancer-susceptibility genes. , 2012, American journal of human genetics.

[13]  Alexander A. Morgan,et al.  Clinical assessment incorporating a personal genome , 2010, The Lancet.

[14]  Patrick F. Chinnery,et al.  Mitochondrial DNA polymerase-γ and human disease , 2006 .

[15]  D. Absher,et al.  Randomized Trial of Personal Genomics for Preventive Cardiology: Design and Challenges , 2012, Circulation. Cardiovascular genetics.

[16]  C. Tyler-Smith,et al.  Deleterious- and disease-allele prevalence in healthy individuals: insights from current predictions, mutation databases, and population-scale resequencing. , 2012, American journal of human genetics.

[17]  Joseph K. Pickrell,et al.  A Systematic Survey of Loss-of-Function Variants in Human Protein-Coding Genes , 2012, Science.

[18]  L Gong,et al.  The Clinical Pharmacogenomics Implementation Consortium: CPIC Guideline for SLCO1B1 and Simvastatin‐Induced Myopathy , 2012, Clinical pharmacology and therapeutics.

[19]  Wei Lu,et al.  Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians , 2011, Nature Genetics.

[20]  P. Radice,et al.  Evidence of a founder mutation of BRCA1 in a highly homogeneous population from southern Italy with breast/ovarian cancer , 2001, Human mutation.

[21]  Rena A. Godfrey,et al.  The National Institutes of Health Undiagnosed Diseases Program: insights into rare diseases , 2011, Genetics in Medicine.

[22]  angesichts der Corona-Pandemie,et al.  UPDATE , 1973, The Lancet.

[23]  J. Shendure,et al.  Exome sequencing as a tool for Mendelian disease gene discovery , 2011, Nature Reviews Genetics.

[24]  Tanya M. Teslovich,et al.  Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes , 2012, Nature Genetics.

[25]  J. Dungan,et al.  Carrier Testing for Severe Childhood Recessive Diseases by Next-Generation Sequencing , 2012 .

[26]  R. Nussbaum,et al.  Genetic/familial high-risk assessment: breast and ovarian. , 2010, Journal of the National Comprehensive Cancer Network : JNCCN.

[27]  C. Croce,et al.  BRCA1 5083del19 Mutant Allele Selectively Up-Regulates Periostin Expression In vitro and In vivo , 2008, Clinical Cancer Research.

[28]  W. Grody,et al.  ACMG recommendations for standards for interpretation and reporting of sequence variations: Revisions 2007 , 2008, Genetics in Medicine.

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

[30]  J. Mega,et al.  Consortium ( CPIC ) guidelines for cytochrome P 450-2 C 19 ( CYP 2 C 19 ) genotype and clopidogrel therapy , 2011 .

[31]  T E Klein,et al.  Clinical Pharmacogenetics Implementation Consortium Guidelines for Thiopurine Methyltransferase Genotype and Thiopurine Dosing: 2013 Update , 2013, Clinical pharmacology and therapeutics.

[32]  J. Mega,et al.  Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C19 Genotype and Clopidogrel Therapy: 2013 Update , 2013, Clinical pharmacology and therapeutics.

[33]  J. Danesh,et al.  Large-scale association analysis identifies new risk loci for coronary artery disease , 2013 .

[34]  Euan A Ashley,et al.  Performance comparison of whole-genome sequencing platforms , 2011, Nature Biotechnology.

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

[36]  P. Shannon,et al.  Exome sequencing identifies the cause of a Mendelian disorder , 2009, Nature Genetics.

[37]  Darlene Riethmaier,et al.  Towards a Universal Clinical Genomics Database: The 2012 International Standards for Cytogenomic Arrays Consortium Meeting , 2013, Human mutation.

[38]  Robert B. Hartlage,et al.  This PDF file includes: Materials and Methods , 2009 .

[39]  I. Tikhonova,et al.  Genetic diagnosis by whole exome capture and massively parallel DNA sequencing , 2009, Proceedings of the National Academy of Sciences.

[40]  S. Gross,et al.  The kappa coefficient of agreement for multiple observers when the number of subjects is small. , 1986, Biometrics.

[41]  F. Couch,et al.  BRCA1 mutations in women attending clinics that evaluate the risk of breast cancer. , 1997, The New England journal of medicine.

[42]  M. Whirl‐Carrillo,et al.  Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C9 and VKORC1 Genotypes and Warfarin Dosing , 2011, Clinical pharmacology and therapeutics.

[43]  P. Stenson,et al.  The Human Gene Mutation Database: 2008 update , 2009, Genome Medicine.

[44]  P. Chinnery,et al.  Mitochondrial DNA polymerase-gamma and human disease. , 2006, Human molecular genetics.

[45]  Omar E. Cornejo,et al.  Phased Whole-Genome Genetic Risk in a Family Quartet Using a Major Allele Reference Sequence , 2011, PLoS genetics.

[46]  P. Stankiewicz,et al.  Whole-genome sequencing in a patient with Charcot-Marie-Tooth neuropathy. , 2010, The New England journal of medicine.

[47]  Kenny Q. Ye,et al.  An integrated map of genetic variation from 1,092 human genomes , 2012, Nature.

[48]  James P Evans,et al.  An informatics approach to analyzing the incidentalome , 2012, Genetics in Medicine.