Clinical Applications of Whole Genome/Whole Exome Sequencing Robert L. Nussbaum, MD, FACMG Division of Genomic Medicine, UCSF AMA – November 11, 2012.

Slides:



Advertisements
Similar presentations
Taking Research and Development to the Clinic: Issues for Physicians AAAS/FDLI Colloquium I Diagnostics and Diagnoses Paths to Personalized Medicine Howard.
Advertisements

ACCF/AHA Clopidogrel Clinical Alert: Approaches to the FDA “Boxed Warning” A Report of the American College of Cardiology Foundation Task Force on Clinical.
Lecture 2 Strachan and Read Chapter 13
Implications of Consanguinity for Routine Diagnostic Testing and Development of Specialist Services Teresa Lamb Clinical Scientist Leeds DNA Laboratory.
CZ5225 Methods in Computational Biology Lecture 9: Pharmacogenetics and individual variation of drug response CZ5225 Methods in Computational Biology.
Genes that affect novelty seeking behavior
Breast Cancer Risk and Risk Assessment Models
Dr. Almut Nebel Dept. of Human Genetics University of the Witwatersrand Johannesburg South Africa Significance of SNPs for human disease.
DNA marker analysis Mrs. Stewart Medical Interventions Central Magnet School.
FDA Panel Comments Adele Schneider, MD, FACMG Victor Center for the Prevention of Jewish Genetic Diseases, Director, Clinical Genetics Albert Einstein.
Computational Tools for Finding and Interpreting Genetic Variations Gabor T. Marth Department of Biology, Boston College
Positional Cloning LOD Sib pairs Chromosome Region Association Study Genetics Genomics Physical Mapping/ Sequencing Candidate Gene Selection/ Polymorphism.
Human Genetics Overview.
Clinical Genotyping and Personalized Medicine Michael D. Kane, PhD (1) Associate Professor of Bioinformatics (2) University Faculty Scholar (3) Chair of.
Microsatellite Instability Detection by Next Generation Sequencing S.J. Salipante, S.M. Scroggins, H.L. Hampel, E.H. Turner, and C.C. Pritchard September.
I inherited What??? You and Your Genes: The Explosive New World of Genetics David Finegold, M.D.
Human Molecular Genetics Section 14–3
XGenCloud. Cloud service intended for automatic interpretation of genetic tests results, allows to form detailed genetic conclusion. Cloud service intended.
Direct-to-Consumer Genetic Testing Developed by Dr. June Carroll, Ms. Shawna Morrison and Dr. Judith Allanson Last updated April 2014.
Doug Brutlag 2011 Genomics & Medicine Doug Brutlag Professor Emeritus of Biochemistry &
Pharmacogenomics and personalized medicines Jean-Marie Boeynaems
University of Utah Department of Human Genetics Pharmacogenomics Louisa A. Stark, Ph.D. Director.
Genomics Alexandra Hayes. Genomics is the study of all the genes in a person, as well as the interactions of those genes with each other and a person’s.
Chapter 12 Human Genetics.
Understanding Genetics of Schizophrenia
Association of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications Mark Bleackley MEDG 505 March.
Genomics and Personalized Care Leming Zhou, PhD Assistant Professor Department of Health Information Management School of Health and Rehabilitation Sciences.
The Human Genome the human genome consists of ~3 billion bp and 30,000-35,000 genes (haploid state) it would fill about 150,000 phone book pages with A’s,
Chapter 13 Carrier Screening. Introduction Carrier screening involves testing of individuals for heterozygosity for genes that would produce significant.
Analyzing DNA Differences PHAR 308 March 2009 Dr. Tim Bloom.
HLA-B*5701 and Abacavir Alec Walker October 2014.
Loss-of-co-Homozygosity mapping and exome sequencing of a Syrian pedigree identified the candidate causal mutation associated with rheumatoid arthritis.
National Heart, Lung, and Blood Institute Women’s Health Initiative Branch Jacques Rossouw, MD Chief, WHI Branch Program for Prevention and Population.
Personalized Medicine The Promise of the Genomic Revolution.
From Genome-Wide Association Studies to Medicine Florian Schmitzberger - CS 374 – 4/28/2009 Stanford University Biomedical Informatics
Personalized Medicine Dr. M. Jawad Hassan. Personalized Medicine Human Genome and SNPs What is personalized medicine? Pharmacogenetics Case study – warfarin.
ABC for the AEA Basic biological concepts for genetic epidemiology Martin Kennedy Department of Pathology Christchurch School of Medicine.
Risk Prediction of Complex Disease David Evans. Genetic Testing and Personalized Medicine Is this possible also in complex diseases? Predictive testing.
Rachel Liao, PhD Coordinator of the Clinical Working Group and the BRCA Challenge demonstration project for the Global Alliance for Genomics and Health.
Lecture-3 EXOME SEQUENCING Huseyin Tombuloglu, Phd GBE423 Genomics & Proteomics.
Paolo Vineis University of Torino and ISI Foundation, Torino, Italy address: GENE-ENVIRONMENT INTERACTIONS IN CANCER.
In The Name of GOD Genetic Polymorphism M.Dianatpour MLD,PHD.
Pharmacogenetics.
Genetic disorders can be due to any of the following factors: A. Monogenetic Disorders: Caused by a mutation in a single gene 1. Autosomal recessive alleles:
Cancer 101: A Cancer Education and Training Program for [Target Population] Date Location Presented by: Presenter 1 Presenter 2.
Regulatory Guidance for Genetic Testing. Three Specific Areas Laboratory tests Results of genetic testing – Clinical – Research GenomeWide Association.
Human Genomics Higher Human Biology. Learning Intentions Explain what is meant by human genomics State that bioinformatics can be used to identify DNA.
Ethics in Clinical Genetics and Genomics Key Knowledge Year 4 Medical Ethics and Law Thread Course, The Ethox Centre, University of Oxford.
EMGO Institute for Health and Care Research Quality of Care Martina Cornel Professor of Community Genetics & Public Health Genomics Genomics in health.
Different microarray applications Rita Holdhus Introduction to microarrays September 2010 microarray.no Aim of lecture: To get some basic knowledge about.
Clinical Department of Psychiatry University of Michigan Medical School Ann Arbor, June 28, 2002 Why Medicine Should be an Information Science Bruce R.
Reliable Identification of Genomic Variants from RNA-seq Data Robert Piskol, Gokul Ramaswami, Jin Billy Li PRESENTED BY GAYATHRI RAJAN VINEELA GANGALAPUDI.
Genome-Wides Association Studies (GWAS) Veryan Codd.
Next generation genomics: translation into clinically useful applications in health care Prof.dr. Martina Cornel
1 Finding disease genes: A challenge for Medicine, Mathematics and Computer Science Andrew Collins, Professor of Genetic Epidemiology and Bioinformatics.
Pharmacogenetics/Pharmacogenomics. Outline Introduction  Differential drug efficacy  People react differently to drugs Why does drug response vary?
Genomics and Disease Gene Identification. Is the Disease Genetic or Environmental.
Hereditary Cancer Predisposition: Updates in Genetic Testing
Interpreting exomes and genomes: a beginner’s guide
Institute of Social and Preventive Medicine, University of Lausanne
Dewey et al. Presented By: Natasha Granneman & Christina Tran
Genomic Analysis: GWAS
PhenGene P2Y12 Test.
An Electronic Medical Record Based Pharmacogenetic Study
Genetic Testing for the Clinician
Interpretation Next Generation Sequencing (Bench Clinic)
Genetic Disorders and Genetic Testing
Content and Labeling of Tests Marketed as Clinical “Whole-Exome Sequencing” Perspectives from a cancer genetics clinician and clinical lab director Allen.
Different mode and types of inheritance
Beyond GWAS Erik Fransen.
Presentation transcript:

Clinical Applications of Whole Genome/Whole Exome Sequencing Robert L. Nussbaum, MD, FACMG Division of Genomic Medicine, UCSF AMA – November 11, 2012

Chair of Genomic Medicine Advisory Board of Complete Genomics, Inc. Conflict of Interest Disclosures

Mythical Scenario A newborn blood spot undergoes whole genome sequencing. It is analyzed for Personal risk for a Mendelian disorder (BRCA1) Pharmacogenetic variants that predict efficacy, side-effects, adverse reactions (CYP2C19 and clopidogrel) Risk for carrying mutations that future children at risk (Ta- Sachs carrier) Tissue-type and Blood type (HLA, ABO) Variants (rare and common) that increase risk for common disorders (CFH and macular degeneration) All the results are recorded in an EMR, communicated to his health care providers, and used to guide health care over the lifespan

Outline Whole Genome and Whole Exome Sequencing Factors Impeding Implementation of WGS/WES sequencing – Limits of the Technology – Limits of Knowledge – Limits of Genetic Determinism

Evaluating A Genetic Test  Patient Sample  Right result from the right patient  Test has predictive value for patient care  Results have value for the patient and doctor  There is value to society in generalizing the testing Analytical Validity Clinical Validity Clinical Utility (“Actionability”) Social Utility

Whole Genome Sequencing (WGS) CLIA ’88 Test Performance Metrics Reportable Range: Portion of the genome from which sequence information can be reliably derived from WGS = ~96.5% Reference Range: Homopolymers, di- and tri-nucleotide repeats, microsatellites Deletions and duplications ~ bp Single nucleotide variants sitting at the end of homopolymers Are outside the typical Reference Range of WGS

Whole Exome Sequencing (WES) by Exon Capture Elute Sequence

~3-5% of Exons, Promoters, Untranslated Regions, and the Bulk of Intron Sequences are not Included in Exome Sequencing Start Stop What Do You Miss With Whole Exome Sequencing? 5’-UTR 3’-UTR

Why Do WES Rather Than WGS? Because you only sequence ~2% of the genome, what you do sequence is covered to tremendous depth You are sequencing the part of the genome we are better at interpreting Current cost of WES is ~$750-$1000 versus $4,000-$10,000 for WGS HOWEVER………

How Good are WES and WGS at Identifying Variants? Because of False Positives, neither approach provides stand-alone “clinical grade” sequencing at the present time and Variants need to be confirmed by conventional sequencing Increases the cost tremendously WES for research = $750 WES for Clinical Use = $8, ,000

Variants in Whole Genome Sequence

“The” Human Genome There is no such thing – there are only Human Genomes There is a “Reference Genome” in databases but it is incomplete Variants are defined as differences from the Reference The more we learn, the more we realize that there are alternative Reference Genomes

Evaluating A Genetic Test  Patient Sample  Right result from the right patient  Test has predictive value for patient care  Results have value for the patient and doctor above and beyond current practice  There is value to society in generalizing the testing Analytical Validity Clinical Validity Clinical Utility (“Actionability”) Social Utility

Clinical Validity Positive Predictive Value Given a + test, how frequently does the patient have, or how frequently will he develop the disease? (“Penetrance”) Negative Predictive Value Given a – test, how frequently is the patient unaffected and will remain so?

The Reason for the Test Matters “Screening” a healthy executive for variants in her DNA that might be of interest Versus “Scanning” a child with a serious disorder for variants in her DNA that might explain the disease and suggest therapy

Genome-Wide Association Studies in Eight Common Diseases SNPS in a Region on Chr 9 are associated with CAD at P <

Palomaki et al. Odds of Developing CAD Depending on 9p21 Genotype

65 year old male No CAD risk factors 40 year old female No CAD risk factors 9p21Genotype 2 Risk 0 Risk Unknown Alleles Alleles 11% 13.2% 9.2% 2% 2.4% 1.7% Risk for Coronary Artery Disease Events over the Next 10 Years Palomaki et al. PPV for 9p21 Genotype for CAD

Combine 13 SNP Loci To Generate Genetic Risk Score for CAD Sipatti et al. A multilocus genetic risk score for coronary heart disease: case-control and prospective cohort analyses, The Lancet Volume 376, Issue 9750, Pages (October 2010)Volume 376, Issue 9750, Pages

Fraction of the Population

Established Common Breast-Cancer Susceptibility Alleles. Pharoah PD et al. N Engl J Med 2008;358:

Pharoah P et al., N Engl J Med 2008; 358: of 10M UK women carry 14 low risk alleles ( %) 7 of 10M UK women carry 14 high risk alleles ( %) Distribution of Genetic Risk in the Population: Seven Breast Cancer Risk Alleles (Avg. risk allele freq. = ~0.35) 20,000 of 10M carry BRCA1/2 mutations Assuming a multiplicative model for interaction between these alleles

Evaluating A Genetic Test  Patient Sample  Right result from the right patient  Test has predictive value for patient care  Results have value for the patient and doctor above and beyond current practice  There is value to society in generalizing the testing Analytical Validity Clinical Validity Clinical Utility (“Actionability”) Social Utility

Clinical Utility of Genetic Testing Explain why a disease occurs Institute preventive measures Anticipate and prevent complications Affect choice of therapy Avoid adverse reactions Determine risk in other family members or in future offspring

Clinical Pharmacogenetics Implementation Consortium Gene-Drug Pairs

Clinical Validity ✔ Clinical Utility ? CYP2C19 genotype was not associated with modification of the effect of clopidogrel on CVD end points or bleeding…Overall there was no significant association of genotype with cardiovascular events Individuals with 1 or more CYP2C19 alleles associated with lower enzyme activity had lower levels of active clopidogrel metabolites less platelet inhibition lower risk of bleeding Individuals with 1 or more CYP2C19 alleles associated with lower enzyme activity had lower levels of active clopidogrel metabolites less platelet inhibition lower risk of bleeding

Actionability: In the Eye of the Beholder

What is “Actionable Information”? How does it differ from Clinical Utility? Information with high Clinical Validity Information that allows a medical decision to be made or therapeutic action to be taken (or not). Founded on evidence (A real problem in genetics where diseases are rare) Information that informs an individual and helps him/her make health decisions

Berg J. et al. Genetics IN Medicine Volume 13, Number 6, June 2011 “Actionability” Rating

Conclusions 1.Genetic Testing is often not straightforward and requires substantial interpretation 2.We do not know how to interpret a lot of genetic information 3.Genetic Testing is not static and what a result means can change over time. WES/WGS only magnify the problems enormously

Barriers to the adoption of pharmacogenetic tests in clinical practice Fragmentation of health-care systems that preclude linking a “lifetime” genetic test result with future medical care (exception: the VA) Limited use of electronic medical records vital to linking test results with medication prescribing/dispensing Health-care systems that do not reward the prevention of disease (or adverse drug effects),

Barriers to the adoption of pharmacogenetic tests in clinical practice Lack of sufficient awareness about genomics on the part of many clinicians, Little of such testing is done preemptively and therefore the results are not available when the prescribing decision is made. Some of these barriers will persist for many years to come.