The Infrastructure for Future Medicine

Slides:



Advertisements
Similar presentations
Genetics and Genomics in Clinical Medicine
Advertisements

The Health Services Researcher of 2020: A Summit on the Future of HSR Data and Methods Learning 2.0: Robust, Rigorous, Relevant, and Rapid Paul Wallace.
Patient Safety and Quality of Care: Role of the Compliance Professional Harvey V. Fineberg, M.D., Ph.D. Sixth Annual National Congress on Health Care Compliance.
Major Cancer Milestones
Regulation of Consumer Tests in California AAAS Meeting June 1-2, 2009 Beatrice OKeefe Acting Chief, Laboratory Field Services California Department of.
Taking Research and Development to the Clinic: Issues for Physicians AAAS/FDLI Colloquium I Diagnostics and Diagnoses Paths to Personalized Medicine Howard.
ACCF/AHA Clopidogrel Clinical Alert: Approaches to the FDA “Boxed Warning” A Report of the American College of Cardiology Foundation Task Force on Clinical.
4 pictures 1 word! Learning Outcomes Examine how future medicine may take into account an individuals genome for personalised medicine Distinguish between.
Genomic Medicine: A Revolution in Medical Practice in the 21 st Century Francis S. Collins, M.D., Ph.D. National Human Genome Research Institute World.
1. 2 Key Applications of Genetic and Genomic Testing (slide 1 of 2) Diagnosis of Disease: Whereby genetic or genomic tests are used to screen a patient.
Implementation Research: Using Science to Guide Implementation of Evidence-Based Practices Brian S. Mittman, PhD Director, VA Center for Implementation.
L1:Apply the concepts of health and wellness to identify health behaviours and factors influencing choice and change in health using an holistic approach.
Wrapup. NHGRI strategic plan What does the NIH think genomics should be for the next 10 years? [Nature, Feb. 2011]
©2007 RALPH SNYDERMAN Third Annual Prospective Health Care Conference "Where are We Now?" Ralph Snyderman, MD Chancellor Emeritus, Duke University November.
Drug Utilization Review (DUR)
16 November 2004Biomedical Imaging BMEN Biomedical Imaging of the Future Alvin T. Yeh Department of Biomedical Engineering Texas A&M University.
1 Biomedical Sciences Public and Environmental Health Regenerative Medicine Translational Research.
Dr. Almut Nebel Dept. of Human Genetics University of the Witwatersrand Johannesburg South Africa Significance of SNPs for human disease.
1 SIMG Florence ppt The Horizons of Predictive Medicine Dr Ian Gilham Worldwide Director, Predictive Medicine Glaxo Wellcome Research and Development.
Bindley Bioscience Center Vision: Nurture interactive communication and interdisciplinary discovery with flexible laboratory project spaces and an open.
Human Genetics Overview.
What Do Toxicologists Do?
Clinical Genotyping and Personalized Medicine Michael D. Kane, PhD (1) Associate Professor of Bioinformatics (2) University Faculty Scholar (3) Chair of.
Introduction of Cancer Molecular Epidemiology Zuo-Feng Zhang, MD, PhD University of California Los Angeles.
Personalized Medicine
Genetic Testing in Genomic Medicine Gail H. Vance M.D. Professor, Department of Medical & Molecular Genetics Indiana University School of Medicine.
Introduction to Molecular Epidemiology Jan Dorman, PhD University of Pittsburgh School of Nursing
The NIH Roadmap for Medical Research
Paola CASTAGNOLI Maria FOTI Microarrays. Applicazioni nella genomica funzionale e nel genotyping DIPARTIMENTO DI BIOTECNOLOGIE E BIOSCIENZE.
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.
Association of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications Mark Bleackley MEDG 505 March.
“Put the Power of Predictive Analytics in the Hands of Clinical Researchers” Filippos Katsampouris Marketing Manager Healthcare & Pharmaceutical Accounts.
BIOLOGIC MARKERS IN OCCUPATIONAL AND ENVIRONMENTAL MEDICINE By: Dr Chavoshi.
Epigenome 1. 2 Background: GWAS Genome-Wide Association Studies 3.
Introduction to Precision Medicine
Pre Med III Genetics Guri Tzivion, PhD Extension 506 Summer 2015 Windsor University School of Medicine.
KEY CONCEPT Biology is the study of all forms of life.
Pharmacogenomics. Developing drugs on the basis of individual genetic differences Tailoring therapies to genetically similar subpopulations results in.
Basma Y. Kentab MSc.. 1. Define ambulatory care 2. Describe the value of ambulatory care practices 3. Explore pharmacy services in some ambulatory care.
Clinical Pharmacy Part 2
Biomedical Research.
Precision Medicine A New Initiative. The Concept of Precision Medicine (PM) The prevention and treatment strategies that take individual variability into.
Pharmacogenetics & Pharmacogenomics Personalized Medicine.
Instruments By Glory Basumata (MSc Applied Genetics) 2011.
Developing medicines for the future and why it is challenging Angela Milne.
Personalized Medicine Dr. M. Jawad Hassan. Personalized Medicine Human Genome and SNPs What is personalized medicine? Pharmacogenetics Case study – warfarin.
Bringing Genomics Home Your DNA: A Blueprint for Better Health Dr. Brad Popovich Chief Scientific Officer Genome British Columbia March 24, 2015 / Vancouver,
Dr Godfrey Grech University of Malta
D4FF55A0-6B6F BF422A9BA9 Present by: Xiao Chen On December 7, 2015.
Pharmacogenetics.
Cancer 101: A Cancer Education and Training Program for [Target Population] Date Location Presented by: Presenter 1 Presenter 2.
Mt. Hood. IOM Report: 10 Years After & More Coming Mitch Greenlick, Ph.D. Oregon State Representative April 21, 2010.
Applying New Science to Drug Safety Janet Woodcock, M.D. Acting Deputy Commissioner for Operations April 15, 2005.
Jan 2002 EDMA The central role of the Medical Laboratory in a World of Managed Health An EDMA presentation of the benefits of in vitro testing as a basis.
Different microarray applications Rita Holdhus Introduction to microarrays September 2010 microarray.no Aim of lecture: To get some basic knowledge about.
Chapter 7: Epidemiology of Chronic Diseases. “The Change You Like to See….” (1 of 3) Chronic diseases result from prolongation of acute illness. – With.
Bringing Genomics Home Your DNA: A Blueprint for Better Health Genome British Columbia November 18, 2015 / West Vancouver, BC.
Doug Brutlag 2011 The impact of genomics on the future of medicine and health Muhammad Faisal 2015-Arid-3638 PhD(scholar) Biochemistry.
1 Copyright © 2012 by Mosby, an imprint of Elsevier Inc. Copyright © 2008 by Mosby, Inc., an affiliate of Elsevier Inc. Chapter 11 Genomics in Public Health.
European Patients’ Academy on Therapeutic Innovation Challenges in Personalised Medicine.
Moiz Bakhiet, MD, PhD, Professor and Chairman
New research areas in personalised medicines
TITIN ANDRI WIHASTUTI SCHOOL OF NURSING FACULTY OF MEDICINE
Biomarkers.
Biomedical Therapies Foundation Standard 1: Academic Foundation
So …What’s the future of medicine?
Pharmacogenomics Genes and Drugs.
BIOSENSOR.
Bio-Rad Overview and Statement of Interests
Presentation transcript:

The Infrastructure for Future Medicine Genomic Medicine: The Infrastructure for Future Medicine TUMS Javad Tavakkoly Bazzaz MD, PhD Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences

Genomic Medicine Vs. Medical Genetics Genetics > Human Genetics > Medical Genetics Genomic Medicine: Understanding variability between individuals allows for more targeted or personalized healthcare based on genetic differences. To link, on a genomic scale, the resources and tools of the Genome Project to major biological processes and systems involved in different diseases.

Systems/Translational Medicine Systems Biology: Understanding the manner in which the parts of an organism interact in complex networks. Systems Medicine: Translation of systems biology into medicine. Translational Genomics: The part of systems medicine that deals with genome-based systems engineering.

Translational Medicine Translation of evidence based research findings into practice. Findings have not been implemented into practice in widespread settings. May take 10 -20 years for original research to translate to routine medical practice.

Goals of Translational Genomics Screen for key genes and gene families that explain specific cellular phenotypes (disease). Use genomic signals to classify/subclassify diseases on a molecular level. Build model networks to study dynamical genome behavior and derive intervention strategies to alter undesirable behavior.

Epigenetics Mechanisms Stable & Dynamic Genomics Epigenetics Mechanisms RNA Interference Gene Expression Histone Modifications DNA Methylation

Translational Research: Bridging the gap between bench and bedside Nature, June 12, 2008

Basic & Applied Sciences! “ There does not exist a category of science to which one can give the name ‘applied science.’ There are science and the applications of science, bound together as the fruit of the tree which bears it.” Louis Pasteur

Genetic Research in Post-Genomic Era Paradigm Shift in Genetic Research in Post-Genomic Era Modified From Peltonen and McKusick, 2001

More Specific Information, Because we know More Specific Information, we can… Diagnose more precisely Provide more effective treatment. Select specific treatment that best fits disease Target the medication to the disorder. Avoid adverse drug reactions. Avoid delay from false starts. Predict risk before symptoms occur Provide earlier treatment. Take preventive action. Manage disease more effectively Eliminate unnecessary treatment. Provide better timing. Adjust treatment as disease changes. AND… AND… AND… Here is what that powerful new individualized knowledge lets us do…. We can diagnose disease much more precisely, which lets us provide more effective treatment. We can select a specific treatment that fits the specific disease of the individual. This allows us to begin treatment promptly, without the false starts of trial and error medicine. It also means we can avoid the adverse drug reactions that drive up deadly complications for patients—along with health costs. We can predict the risk of a disease well before symptoms occur, which lets us take preventive and therapeutic action earlier. And we are better able to manage the condition over time, as it changes and as the patient’s needs change. Today, we will look at examples of all of these. AND…

Tools Needed for Prediction & Personalized Care Decision Support Tools: Monitor Progression Predict Events Inform Therapeutics Assess Risk Refine Assessment Predict/Diagnose Baseline Risk Initiating Events Earliest Molecular Detection Earliest Clinical Detection Typical Current Intervention 1/reversibility Cost Disease Burden Time Baseline Risk Preclinical Progression Disease Initiation and Progression Stable Genomics: (SNPs) Haplotype Mapping , Gene Sequencing Dynamic Genomics: Gene Expression Proteomics Metabolomics Molecular Imaging Therapeutic Decision Support Sources of New Biomarkers:

Genetics is becoming a driving force in medical decision making Preparing for the Age of Genomic Medicine Genetics is becoming a driving force in medical decision making

Uses of Genetic Information Development of gene therapy Genetic testing for risk stratification Use of genetic information for treatment decisions & drug development

Medicine of the past could be called “trial-and-error” medicine Observe Diagnose Treat Monitor response Adjust Let’s begin by examining what medicine has looked like in the past. Think of it as trial-and-error medicine. A physician observes the patient’s condition through a physical exam, asks about medical history, and makes a “most likely” diagnosis based on the knowledge and evidence that is available. The physician then decides upon the treatment that he or she believes is most appropriate. Then the physician monitors the response to decide whether to stay the course or try an alternative. Trial-and-error medicine represents a vast improvement over what came before—largely the “guesswork” of the 19th and early 20th centuries. And it often works.

Interindividual Variability in Drug Response Disease Drug Class Rate of Poor Response Asthma Beta-agonists 40-75% Hypertension Various 30% Solid Cancers Various 70% Depression SSRIs, tricyclics 20-40% Diabetes Sulfonylureas, others 50% Arthritis NSAIDs, COX-2 inhibitors 30-60% Schizophrenia Various 25-75%

Factors Contributing to Interindividual Variability in Drug Disposition and Action Age Race/ethnicity Weight Gender Concomitant Diseases Concomitant Drugs Social factors GENETICS PERSONALIZED MEDICINE

Diagnose more precisely Genetic tests identify DNA mutations of childhood leukemia, enabling physicians to choose the treatment that fits it precisely. The impact of genetic tests and genome-based cancer drugs on survival of childhood leukemia Acute lymphoblastic leukemia is most common form of childhood leukemia Genetic tests identify subtypes; allow precise treatment and timing Today’s cure-rate exceeds 80% vs. 4% in the 1960s One of the best examples of diagnosing disease more effectively involves the most common form of childhood leukemia—acute lymphoblastic leukemia. Genetic tests are able to determine the exact chromosomal structure of the individual child’s disease. That allows physicians to select the optimal drug and dosage for each child. You can see some of the results: In 1962, about 4 percent of children were cured. Now that is over 80 percent. Source: New England Journal of Medicine, 2006, 200l; Personalized Medicine Coalition, 2006.

Today, Cancer is experiencing a shift toward precision medicine Farber develops 1st chemotherapy for leukemia Novartis launches Gleevec, the 1st molecular targeted drug, to treat myeloid leukemia 2 types: leukemia & lymphoma 1920 1930 1940 1950 1960 1970 1980 1990 2000 2010 Disease of the blood 38 types of leukemia; 51 types of lymphoma 3 types of leukemia (acute, chronic, preleukemia) and 2 types of lymphoma (indolent, aggressive) A deeper understanding of molecular biology and the human genome is enabling scientists and clinicians to begin diagnosing and treating cancers based on their molecular characteristics rather than just gross anatomical observations Source: Mara Aspinall, Genzyme 18

Main health-related challenges Cardiovascular Cancer Diabetes-obesity Undernutrition/malnutrition Infectious diseases Allergies Ageing process Complex biological problems! Polygenic + environmental effects!

NCRR Strategic Plan 2009–2013 Translating Research from Basic Discovery to Improved Patient Care

NIH Roadmap Approach to accelerate fundamental discovery and translation of research knowledge into effective prevention strategies and new treatments. Strategic initiatives will address critical roadblocks and knowledge gaps that currently constrain rapid progress in biomedical research. Synergize the work of many NIH Institutes and Centers. http://nihroadmap.nih.gov

7 common barriers to translation Basic research not clinically relevant ( generated for its own sake!) Clinical practice guidelines in literature insufficient for translation. Comparisons among programs hampered by competition, conflict of interest. Perception that translation will be lengthy and expensive Time necessary to train researchers to communicate and share across fields Practitioners unable to utilize an innovation Researchers don’t interpret findings so that they can’t be easily disseminated

Technology Platforms Patient Benefits

Biomarker A biomarker, or biological marker, is in general a substance used as an indicator of a biological state. It is a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. It is used in many scientific fields. In medicine, a biomarker can be a traceable substance that is introduced into an organism as a means to examine organ function or other aspects of health.. It can also be a substance whose detection indicates a particular disease state, for example, the presence of an antibody may indicate an infection. More specifically, a biomarker indicates a change in expression or state of a protein that correlates with the risk or progression of a disease, or with the susceptibility of the disease to a given treatment. A useful way of finding genetic causes for diseases such as schizophrenia has been the use of a special kind of biomarker called an endophenotype.

Biosensor A biosensor is an analytical device for the detection of an analyte that combines a biological component with a physicochemical detector component. An analyte is a substance or chemical constituent that is determined in an analytical procedure, such as a titration. For instance, in an immunoassay, the analyte may be the ligand or the binder, while in blood glucose testing, the analyte is glucose. In medicine, analyte often refers to the type of test being run on a patient, as the test is usually determining a chemical substance in the human body. An analyte itself cannot be measured, but a measurable property of the analyte can. For instance, one cannot measure a table (analyte- component) but, the height, width, etc. of a table can be measured. Likewise, one cannot measure glucose but can measure the glucose concentration. In this example "glucose" is the component and "concentration" is the kind-of-property. Analyte

Biosensor A biosensor consists of 3 parts: the sensitive biological element (biological material -e.g. tissue, microorganisms, organelles, cell receptors, enzymes, antibodies, nucleic acids, etc.-, a biologically derived material or biomimic). the transducer or the detector element (works in a physicochemical way; optical, piezoelectric, electrochemical, etc.) that transforms the signal resulting from the interaction of the analyte with the biological element into another signal (i.e., transducers) that can be more easily measured and quantified; Associated electronics or signal processors that are primarily responsible for the display of the results in a user-friendly way. A common example of a commercial biosensor is the blood glucose biosensor, which uses the enzyme glucose oxidase to break blood glucose down. In doing so it first oxidizes glucose and uses two electrons to reduce the FAD (a component of the enzyme) to FADH2. This in turn is oxidized by the electrode (accepting two electrons from the electrode) in a number of steps. The resulting current is a measure of the concentration of glucose. In this case, the electrode is the transducer and the enzyme is the biologically active component.

Predict Risk of Disease Before Symptoms Genetic tests identify variations in the BRCA 1 and BRCA 2 genes that increase risks for breast and ovarian cancer. Genetic tests identify greatly increased hereditary risk for breast and ovarian cancer Knowledge of increased risk allows preventive measures, such as closer monitoring, risk avoidance, and preventive surgery or chemotherapy. Lifetime risk of developing breast cancer… …with BRCA 1 and 2 = 50% - 85% ….without = 13% Genetic tests are also able to predict a patient’s risk well before symptoms appear. This is illustrated well in the case of the BRCA1 and BRCA2 genes that increase a patient’s risk for breast and ovarian cancer. According to estimates of lifetime risk, about 13.2 percent of women in the general population will develop breast cancer, compared with estimates of 36 – 85 percent of women with an altered BRCA1 or BRCA2 gene. In other words, women with these gene alterations are 3 – 7 times more likely to develop breast cancer than women without. You can also see that these genes dramatically increase the risks of developing ovarian cancer as well. This information empowers patients and physicians by knowing what they are facining and having the option to take action that can lessen the risk. Closer monitoring Earlier treatment through surgery or preventive chemotherapy Greater risk-avoidance Lifetime risk of developing ovarian cancer… …with BRCA 1 and 2 = 10% - 45% …without = 1.7%

Managing Warfarine Dosing Warfarin is metabolized primarily via oxidation in the liver by CYP2C9, and exerts its anticoagulant effect by inhibiting the protein vitamin K epoxide reductase complex, subunit 1 (VKORC1). Three single nucleotide polymorphisms (SNPs), two in the CYP2C9 gene and one in the VKORC1 gene, have been found to play key roles in determining the effect of warfarin therapy on coagulation. Variants of two genes, CYP2C9 and VKORC1 account for 30-50% of the variability.

Predicted economic benefit of CYP2C9 testing for warfarin dosing Predict 1 major bleed prevented for every 44 patients screened ($135/assay) $6,000 testing costs ~ cost of 1 major bleed Neutral economic result, but significant improvement in patient outcome Prospective trial needed Higashi and Veenstra, Am J Manag Care 2003; 9: 493-500

Using Genetic Information to Predict Drug Metabolism: The AmpliChip CYP450 A range of drug metabolism phenotypes is observed for individuals based upon the particular cytochrome P-450 genes they possess. Many drugs metabolized through cytochrome P-450 pathway. Individual variability in CYP450 isoforms contributes to observed range of metabolic phenotypes. Analyzes genetic variation in CYP450-CYP2D6 and CYP450-CYP2D19 genes approved for use in U.S. and Europe Many drugs metabolized through cytochrome P-450 pathway Individual variability in CYP450 isoforms contributes to observed range of metabolic phenotypes. Analyzes genetic variation in CYP450-CYP2D6 and CYP450-CYP2D19 genes Approved for use in U.S. and Europe Source: Caraco, Y., N Engl J Med, 2004

Genomic medicine implies personalization and all its benefits Diagnosis/ predicting risk of disease Determining whether a treatment is working Monitoring healthy people to detect early signs of disease Producing safer drugs by predicting potential for adverse effects earlier Producing better medical products: Producing new classes of structural materials that are expected to bring about lighter, stronger, smarter, cheaper, cleaner, and more precise medical products Producing better medical products Ready access to information Decreasing health care costs 31

Genetic Testing Where will it be used? Diagnosis Prognosis Pharmaceutical Development Therapeutic Selection & Monitoring Screening & Risk Assessment Prevention Genetic testing will impact nearly every clinical decision health care providers make

Genetic Testing Market Number of Available Tests* 1000 1000 945 862 811 800 667 625 600 485 420 400 303 200 Major Point Graph does not include tests for hereditary cancer and infectious disease. This is important because most of the audience members will think of genetic testing as that class of tests for the rare single gene disorders. The point here is to open their eyes to the fact that genetics is becoming a greater part of everyday medicine. The number of tests reported to GeneTests as of 7/31/02 was 943. We are using 1,000 as an estimate of all of 2002. GeneTests is dependent upon self-reporting by the laboratories; therefore, does not capture all the genetic tests actually available. 2003 1995 1996 1997 1998 1999 2000 2001 2002 * Excludes genetic tests for hereditary cancers and infectious diseases Source: GeneTests.org

Genetic Testing Market Growth in Annual Dollars Largest dollar growth in cancer tests (41% of Total by 2006) 1000 Cancer (257% growth) Prenatal (46% growth) 800 Predisposition (254% growth) 600 Million $ Total (134% growth) 400 Additional Information Total genetic testing industry projected to grow from $374.9M to $877.2M Cancer projected to grow from $101.3 to $361.3 Prenatal projected to grow from $217.5 to $316.7 Predisposition (e.g. Huntington’s, Alzheimer projected to grow from $56.2 to $199.2 These numbers do not include genetic testing for infectious disease. 200 2001 2002 2003 2004 2005 2006 Source: Frost and Sullivan, IVD Technology Nov/Dec 2001

Why Do Health Professionals Need to Prepare for Genomic Medicine? Need to learn to “think genetically” - to: realize when genetic factors play a role effectively use family hx & genetic tests be able to explain genetics concepts deal with “risk” & genetic predisposition realize personal and societal impact of genetic information protect genetic privacy use genetics to individualize patient care use genetics to preserve health

“If it were not for the great variability among individuals, medicine might as well be a science and not an art” Sir William Osler, 1892 36

Thank You!