1
|
Khoury MJ. From Genes to Public Health: The Journey Continues! Public Health Genomics 2025; 28:144-149. [PMID: 40311604 DOI: 10.1159/000545406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/18/2025] [Indexed: 05/03/2025] Open
Affiliation(s)
- Muin J Khoury
- Department of Epidemiology, Rollins, School of Public Health, Atlanta, Georgia, USA
- Public Health Genetics Institute, University of Washington, School of Public Health, Seattle, Washington, USA
| |
Collapse
|
2
|
Head M, Cohn B, Wernli KJ, Palazzo L, Ehrlich K, Matson A, Knerr S. Young Women's Perspectives on Being Screened for Hereditary Breast and Ovarian Cancer Risk During Routine Primary Care. Womens Health Issues 2024; 34:268-275. [PMID: 38448251 PMCID: PMC11116046 DOI: 10.1016/j.whi.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE The U.S. Preventive Services Task Force recommends screening women to identify individuals eligible for genetic counseling based on a priori hereditary breast and ovarian cancer syndrome (HBOC) risk (i.e., risk assessment). However, risk assessment has not been widely integrated into primary care. This qualitative study explored young women's views on implementing routine HBOC risk assessment with a focus on equity and patient-centeredness. METHODS We conducted group discussions with young women (aged 21-40 years) receiving care in an integrated health care system. Discussion groups occurred in two phases and used a modified deliberative approach that included a didactic component and prioritized developing consensus. Twenty women participated in one of three initial small group discussions (phase one). All 20 were invited to participate in a subsequent large group discussion (phase two), and 15 of them attended. FINDINGS Key themes and recommendations were as follows. Risk assessment should be accessible, contextualized, and destigmatized to encourage participation and reduce anxiety, particularly for women who do not know their family history. Providers conducting risk assessments must be equipped to address women's informational needs, relieve emotionality, and plan next steps after positive screens. Finally, to minimize differential screening uptake, health care systems must prioritize equity in program design and contribute to external educational and outreach efforts. CONCLUSION Young women see pragmatic opportunities for health systems to optimize HBOC screening implementation.
Collapse
Affiliation(s)
- Mady Head
- Genetic Counseling Graduate Program, School of Medicine, University of Washington, Seattle, Washington
| | - Betty Cohn
- Institute for Public Health Genetics, School of Public Health, University of Washington, Seattle, Washington
| | - Karen J Wernli
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Lorella Palazzo
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Kelly Ehrlich
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Abigail Matson
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Sarah Knerr
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, Washington.
| |
Collapse
|
3
|
Wildin RS. Cost Effectiveness of Genomic Population Health Screening in Adults: A Review of Modeling Studies and Future Directions. J Appl Lab Med 2024; 9:92-103. [PMID: 38167759 DOI: 10.1093/jalm/jfad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Detecting actionable health risks for genetic diseases prior to symptomatic presentation at population scale using genomic test technologies is a preventive health innovation being piloted in multiple locations. Standard practice is to screen for risks only in those with personal or family history of specific disease. Genomic population heath screening has proven feasible and potentially scalable. The value of this intervention in terms of economic benefit has been scientifically modeled by several groups. CONTENT Eight recent cost-effectiveness modeling studies for high penetrance monogenic dominant diseases that used input parameters from 3 different countries are reviewed. Results and their uses in refining implementations are analyzed and the roles for laboratory medicine in facilitating success are discussed. SUMMARY The reviewed studies generally found evidence for cost-effectiveness of genomic population health screening in at least a subset of their base case screening scenario. Sensitivity analyses identified opportunities for improving the likelihood of cost-effectiveness. On the whole, the modeling results suggest genomic population health screening is likely to be cost-effective for high penetrance disorders in younger adults, especially with achievable reductions in test cost effected partially through combining tests for individual disorders into one screening procedure. Policies founded on the models studied should consider limitations of the modeling methods and the potential for impacts on equity and access in the design and implementation of genomic screening programs.
Collapse
Affiliation(s)
- Robert S Wildin
- Departments of Pathology & Laboratory Medicine and Pediatrics, The Larner College of Medicine at the University of Vermont, Burlington, VT, United States
| |
Collapse
|
4
|
Allen CG, McBride CM, Escoffery C, Guan Y, Hood C, Zaho J, Brody G, An W. Developing and assessing a kin keeping scale with application to identifying central influencers in African American family networks. J Community Genet 2023; 14:593-603. [PMID: 37648941 PMCID: PMC10725405 DOI: 10.1007/s12687-023-00665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
Promoting family communication about inherited disease risk is an arena in which family systems theory is highly relevant. One family systems' construct that can support promotion of family communication regarding inherited disease risk is the notion of "kin keeping." However, kin keeping and whether it might be capitalized on to encourage family communication about inherited risk has been understudied. The goal of this report was to propose a broadened conceptualization of kin keeping that distinguishes between a structural functional perspective (role conceptualization) and transitional behaviors (skill conceptualization), and to develop and evaluate a scale that would enable this assertion to be tested among a sample of African American community health workers. We developed a scale using four steps: item development using concept analysis and content validity, scale development among a national sample (n = 312), scale evaluation using exploratory factor analysis (n = 52), and scale reduction. We then posed suppositions of associations that would indicate whether the developed kin keeping measure was assessing a specific family role or set of behaviors. Our results included the development of the first quantitative measure of kin keeping (9- and 15-item scales). Model fit for 9-item scale (CFI = 0.97, AFGI = 0.89, RMSEA = 0.09, SMRM = 0.06) and model fit for 15-item scale (CFI = 0.97, AFGI = 0.89, RMSEA = 0.06, SMRM = 0.05). These findings allow us to move toward more rigorous research about the role of kin keeping on information sharing and health decision making. Results also suggest that, contrary to the historical structural functional conceptualization of kin keeping as a role, kin keeping might also be conceptualized as a behavior or set of modifiable behaviors. Ultimately, the kin keeping scale could be used to operationalize kin keeping in various theoretical models and frameworks, guide intervention development to encourage or train for kin keeping behaviors, and test assumptions of whether families vary in the density of kin keeping.
Collapse
Affiliation(s)
- Caitlin G Allen
- Medical University of South Carolina, Charleston, SC, USA.
- Emory University, Atlanta, GA, USA.
| | | | | | - Yue Guan
- Emory University, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
5
|
Li C, Pan Y, Zhang R, Huang Z, Li D, Han Y, Larkin C, Rao V, Sun X, Kelly TN. Genomic Innovation in Early Life Cardiovascular Disease Prevention and Treatment. Circ Res 2023; 132:1628-1647. [PMID: 37289909 PMCID: PMC10328558 DOI: 10.1161/circresaha.123.321999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality globally. Although CVD events do not typically manifest until older adulthood, CVD develops gradually across the life-course, beginning with the elevation of risk factors observed as early as childhood or adolescence and the emergence of subclinical disease that can occur in young adulthood or midlife. Genomic background, which is determined at zygote formation, is among the earliest risk factors for CVD. With major advances in molecular technology, including the emergence of gene-editing techniques, along with deep whole-genome sequencing and high-throughput array-based genotyping, scientists now have the opportunity to not only discover genomic mechanisms underlying CVD but use this knowledge for the life-course prevention and treatment of these conditions. The current review focuses on innovations in the field of genomics and their applications to monogenic and polygenic CVD prevention and treatment. With respect to monogenic CVD, we discuss how the emergence of whole-genome sequencing technology has accelerated the discovery of disease-causing variants, allowing comprehensive screening and early, aggressive CVD mitigation strategies in patients and their families. We further describe advances in gene editing technology, which might soon make possible cures for CVD conditions once thought untreatable. In relation to polygenic CVD, we focus on recent innovations that leverage findings of genome-wide association studies to identify druggable gene targets and develop predictive genomic models of disease, which are already facilitating breakthroughs in the life-course treatment and prevention of CVD. Gaps in current research and future directions of genomics studies are also discussed. In aggregate, we hope to underline the value of leveraging genomics and broader multiomics information for characterizing CVD conditions, work which promises to expand precision approaches for the life-course prevention and treatment of CVD.
Collapse
Affiliation(s)
- Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Yang Pan
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Davey Li
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Yunan Han
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Claire Larkin
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Varun Rao
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| |
Collapse
|
6
|
Guzauskas GF, Garbett S, Zhou Z, Schildcrout JS, Graves JA, Williams MS, Hao J, Jones LK, Spencer SJ, Jiang S, Veenstra DL, Peterson JF. Population Genomic Screening for Three Common Hereditary Conditions : A Cost-Effectiveness Analysis. Ann Intern Med 2023; 176:585-595. [PMID: 37155986 PMCID: PMC11791829 DOI: 10.7326/m22-0846] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND The cost-effectiveness of screening the U.S. population for Centers for Disease Control and Prevention (CDC) Tier 1 genomic conditions is unknown. OBJECTIVE To estimate the cost-effectiveness of simultaneous genomic screening for Lynch syndrome (LS), hereditary breast and ovarian cancer syndrome (HBOC), and familial hypercholesterolemia (FH). DESIGN Decision analytic Markov model. DATA SOURCES Published literature. TARGET POPULATION Separate age-based cohorts (ages 20 to 60 years at time of screening) of racially and ethnically representative U.S. adults. TIME HORIZON Lifetime. PERSPECTIVE U.S. health care payer. INTERVENTION Population genomic screening using clinical sequencing with a restricted panel of high-evidence genes, cascade testing of first-degree relatives, and recommended preventive interventions for identified probands. OUTCOME MEASURES Incident breast, ovarian, and colorectal cancer cases; incident cardiovascular events; quality-adjusted survival; and costs. RESULTS OF BASE-CASE ANALYSIS Screening 100 000 unselected 30-year-olds resulted in 101 (95% uncertainty interval [UI], 77 to 127) fewer overall cancer cases and 15 (95% UI, 4 to 28) fewer cardiovascular events and an increase of 495 quality-adjusted life-years (QALYs) (95% UI, 401 to 757) at an incremental cost of $33.9 million (95% UI, $27.0 million to $41.1 million). The incremental cost-effectiveness ratio was $68 600 per QALY gained (95% UI, $41 800 to $88 900). RESULTS OF SENSITIVITY ANALYSIS Screening 30-, 40-, and 50-year-old cohorts was cost-effective in 99%, 88%, and 19% of probabilistic simulations, respectively, at a $100 000-per-QALY threshold. The test costs at which screening 30-, 40-, and 50-year-olds reached the $100 000-per-QALY threshold were $413, $290, and $166, respectively. Variant prevalence and adherence to preventive interventions were also highly influential parameters. LIMITATIONS Population averages for model inputs, which were derived predominantly from European populations, vary across ancestries and health care environments. CONCLUSION Population genomic screening with a restricted panel of high-evidence genes associated with 3 CDC Tier 1 conditions is likely to be cost-effective in U.S. adults younger than 40 years if the testing cost is relatively low and probands have access to preventive interventions. PRIMARY FUNDING SOURCE National Human Genome Research Institute.
Collapse
Affiliation(s)
- Gregory F. Guzauskas
- The CHOICE Institute, Department of Pharmacy, University of Washington, Seattle, Washington
| | - Shawn Garbett
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Zilu Zhou
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - John A. Graves
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Jing Hao
- Department of Genomic Health and Department of Population Health Sciences, Geisinger, Danville, Pennsylvania
| | - Laney K. Jones
- Department of Population Health Sciences and Heart Institute, Geisinger, Danville, Pennsylvania
| | - Scott J. Spencer
- Institute for Public Health Genetics, University of Washington, Seattle, Washington
| | - Shangqing Jiang
- The CHOICE Institute, Department of Pharmacy, University of Washington, Seattle, Washington
| | - David L. Veenstra
- The CHOICE Institute, Department of Pharmacy, and Institute for Public Health Genetics, University of Washington, Seattle, Washington
| | - Josh F. Peterson
- Department of Biomedical Informatics and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
7
|
Wang C, Lu H, Bowen DJ, Xuan Z. Implementing digital systems to facilitate genetic testing for hereditary cancer syndromes: An observational study of 4 clinical workflows. Genet Med 2023; 25:100802. [PMID: 36906849 DOI: 10.1016/j.gim.2023.100802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
PURPOSE National efforts have prioritized the identification of effective methods for increasing case ascertainment and delivery of evidence-based health care for individuals at elevated risk for hereditary cancers. METHODS This study examined the uptake of genetic counseling and testing following the use of a digital cancer genetic risk assessment program implemented at 27 health care sites in 10 states using 1 of 4 clinical workflows: (1) traditional referral, (2) point-of-care scheduling, (3) point-of-care counseling/telegenetics, and (4) point-of-care testing. RESULTS In 2019, 102,542 patients were screened and 33,113 (32%) were identified as at high risk and meeting National Comprehensive Cancer Network genetic testing criteria for hereditary breast and ovarian cancer, Lynch syndrome, or both. Among those identified at high risk, 5147 (16%) proceeded with genetic testing. Genetic counseling uptake was 11% among the sites with workflows that included seeing a genetic counselor before testing, with 88% of patients proceeding with genetic testing after counseling. Uptake of genetic testing across sites varied significantly by clinical workflow (6% referral, 10% point-of-care scheduling, 14% point-of-care counseling/telegenetics, and 35% point-of-care testing, P < .0001). CONCLUSION Study findings highlight the potential heterogeneity of effectiveness attributable to different care delivery approaches for implementing digital hereditary cancer risk screening programs.
Collapse
Affiliation(s)
- Catharine Wang
- Department of Community Health Sciences, Boston University School of Public Health, Boston, MA.
| | | | - Deborah J Bowen
- Department of Bioethics and Humanities, School of Public Health, University of Washington, Seattle, WA
| | - Ziming Xuan
- Department of Community Health Sciences, Boston University School of Public Health, Boston, MA
| |
Collapse
|
8
|
Wildin RS, Gerrard DL, Leonard DGB. Real-World Results from Combined Screening for Monogenic Genomic Health Risks and Reproductive Risks in 300 Adults. J Pers Med 2022; 12:jpm12121962. [PMID: 36556183 PMCID: PMC9782229 DOI: 10.3390/jpm12121962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
New methods and demonstrations of feasibility guide future implementation of genomic population health screening programs. This is the first report of genomic population screening in a primary care, non-research setting using existing large carrier and health risk gene sequencing panels combined into one 432-gene test that is offered to adults of any health status. This report summarizes basic demographic data and analyses patterns of pathogenic and likely pathogenic genetic findings for the first 300 individuals tested in this real-world scenario. We devised a classification system for gene results to facilitate clear message development for our Genomic Medicine Action Plan messaging tool used to summarize and activate results for patients and primary care providers. Potential genetic health risks of various magnitudes for a broad range of disorders were identified in 16% to 34% of tested individuals. The frequency depends on criteria used for the type and penetrance of risk. 86% of individuals are carriers for one or more recessive diseases. Detecting, reporting, and guiding response to diverse genetic health risks and recessive carrier states in a single primary care genomic screening test appears feasible and effective. This is an important step toward exploring an exome or genome sequence as a multi-purpose clinical screening tool.
Collapse
Affiliation(s)
- Robert S. Wildin
- Laboratory Medicine and Pediatrics & Departments of Pathology, Robert Larner M.D. College of Medicine at the University of Vermont, University of Vermont Health Network, Burlington, VT 05401, USA
- Correspondence:
| | - Diana L. Gerrard
- Laboratory Medicine & Department of Pathology, University of Vermont Medical Center, Burlington, VT 05401, USA
| | - Debra G. B. Leonard
- Laboratory Medicine & Department of Pathology, Robert Larner M.D. College of Medicine at the University of Vermont, University of Vermont Health Network, Burlington, VT 05401, USA
| |
Collapse
|
9
|
Nurses’ Knowledge, Attitudes, Confidence, and Practices with Genetics and Genomics: A Theory-Informed Integrative Review Protocol. J Pers Med 2022; 12:jpm12091358. [PMID: 36143143 PMCID: PMC9505976 DOI: 10.3390/jpm12091358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction: As key healthcare providers, nurses require genomic competency to fulfil their professional obligations in the genomic era. Prior research suggests that nurses have limited competency with genomics-informed practice. Concepts in the Rogers’ Diffusion of Innovation (DOI) theory (i.e., knowledge, attitudes, and attributes of innovation adopters) provide a framework to understand the process of adoption of innovations, such as genomics, across organizations. We aim to synthesize what is known about the adoption of genomics across nursing within the DOI framework to identify gaps and opportunities to enact sustained adoption of genomics in nursing. Methods and analysis: An integrative literature review, following Whittemore and Knafl’s five steps, will be conducted to evaluate qualitative, quantitative, and mixed-method primary studies that meet inclusion and exclusion criteria. The MEDLINE, PsychINFO, CINAHL, Cochrane, and Sociological Abstracts electronic databases will be searched in addition to the ancestry search method. Two researchers will perform independent screening of studies, quality appraisal using the Mixed-Methods Appraisal Tool, and data analysis using the narrative synthesis method. Disagreements will be resolved by a third reviewer. Findings in this review could be used to develop theory- and evidence-informed strategies to support the sustained adoption of genomics in nursing.
Collapse
|
10
|
Khoury MJ, Bowen S, Dotson WD, Drzymalla E, Green RF, Goldstein R, Kolor K, Liburd LC, Sperling LS, Bunnell R. Health equity in the implementation of genomics and precision medicine: A public health imperative. Genet Med 2022; 24:1630-1639. [PMID: 35482015 PMCID: PMC9378460 DOI: 10.1016/j.gim.2022.04.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022] Open
Abstract
Recent reviews have emphasized the need for a health equity agenda in genomics research. To ensure that genomic discoveries can lead to improved health outcomes for all segments of the population, a health equity agenda needs to go beyond research studies. Advances in genomics and precision medicine have led to an increasing number of evidence-based applications that can reduce morbidity and mortality for millions of people (tier 1). Studies have shown lower implementation rates for selected diseases with tier 1 applications (familial hypercholesterolemia, Lynch syndrome, hereditary breast and ovarian cancer) among racial and ethnic minority groups, rural communities, uninsured or underinsured people, and those with lower education and income. We make the case that a public health agenda is needed to address disparities in implementation of genomics and precision medicine. Public health actions can be centered on population-specific needs and outcomes assessment, policy and evidence development, and assurance of delivery of effective and ethical interventions. Crucial public health activities also include engaging communities, building coalitions, improving genetic health literacy, and building a diverse workforce. Without concerted public health action, further advances in genomics with potentially broad applications could lead to further widening of health disparities in the next decade.
Collapse
Affiliation(s)
- Muin J Khoury
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA.
| | - Scott Bowen
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - W David Dotson
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Emily Drzymalla
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Ridgely F Green
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Robert Goldstein
- Office of the Associate Director of Policy and Strategy, Centers for Disease Control and Prevention, Atlanta, GA; Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Katherine Kolor
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Leandris C Liburd
- Office of Minority Health and Health Equity, Centers for Disease Control and Prevention, Atlanta, GA
| | | | - Rebecca Bunnell
- Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| |
Collapse
|
11
|
Salikhanov I, Heinimann K, Chappuis P, Buerki N, Graffeo R, Heinzelmann V, Rabaglio M, Taborelli M, Wieser S, Katapodi MC. Swiss cost-effectiveness analysis of universal screening for Lynch syndrome of patients with colorectal cancer followed by cascade genetic testing of relatives. J Med Genet 2021; 59:924-930. [PMID: 34782441 PMCID: PMC9411888 DOI: 10.1136/jmedgenet-2021-108062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/04/2021] [Indexed: 12/25/2022]
Abstract
Background We estimated the cost-effectiveness of universal DNA screening for Lynch syndrome (LS) among newly diagnosed patients with colorectal cancer (CRC) followed by cascade screening of relatives from the Swiss healthcare system perspective. Methods We integrated decision trees with Markov models to calculate incremental cost per quality-adjusted life-year saved by screening all patients with CRC (alternative strategy) compared with CRC tumour-based testing followed by DNA sequencing (current strategy). Results The alternative strategy has an incremental cost-effectiveness ratio of CHF65 058 compared with the current strategy, which is cost-effective according to Swiss standards. Based on annual incidence of CRC in Switzerland, universal DNA screening correctly identifies all 123 patients with CRC with LS, prevents 17 LS deaths and avoids 19 CRC cases, while the current strategy leads to 32 false negative results and 253 LS cases lost to follow-up. One way and probabilistic sensitivity analyses showed that universal DNA testing is cost-effective in around 80% of scenarios, and that the cost of DNA testing and the number of invited relatives per LS case determine the cost-effectiveness ratio. Conclusion Results can inform policymakers, healthcare providers and insurance companies about the costs and benefits associated with universal screening for LS and cascade genetic testing of relatives.
Collapse
Affiliation(s)
- Islam Salikhanov
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Karl Heinimann
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, BS, Switzerland
| | - Pierre Chappuis
- Oncogenetics Unit, Division of Oncology, Division of Genetic Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Nicole Buerki
- Women's Clinic, Basel University Hospital, Basel, Switzerland
| | - Rossella Graffeo
- Breast Unit of Southern Switzerland (CSSI), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | | - Manuela Rabaglio
- Medical Oncology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Monica Taborelli
- Genetic Services, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Simon Wieser
- Winterthur Institute of Health Economics, Zurich University of Applied Sciences, Winterthur, Switzerland
| | - Maria C Katapodi
- Department of Clinical Research, University of Basel, Basel, Switzerland
| |
Collapse
|
12
|
Penetrance and outcomes at 1-year following return of actionable variants identified by genome sequencing. Genet Med 2021; 23:1192-1201. [PMID: 33824501 PMCID: PMC9839314 DOI: 10.1038/s41436-021-01142-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 01/17/2023] Open
Abstract
PURPOSE We estimated penetrance of actionable genetic variants and assessed near-term outcomes following return of results (RoR). METHODS Participants (n = 2,535) with hypercholesterolemia and/or colon polyps underwent targeted sequencing of 68 genes and 14 single-nucleotide variants. Penetrance was estimated based on presence of relevant traits in the electronic health record (EHR). Outcomes occurring within 1-year of RoR were ascertained by EHR review. Analyses were stratified by tier 1 and non-tier 1 disorders. RESULTS Actionable findings were present in 122 individuals and results were disclosed to 98. The average penetrance for tier 1 disorder variants (67%; n = 58 individuals) was higher than in non-tier 1 variants (46.5%; n = 58 individuals). After excluding 45 individuals (decedents, nonresponders, known genetic diagnoses, mosaicism), ≥1 outcomes were noted in 83% of 77 participants following RoR; 78% had a process outcome (referral to a specialist, new testing, surveillance initiated); 68% had an intermediate outcome (new test finding or diagnosis); 19% had a clinical outcome (therapy modified, risk reduction surgery). Risk reduction surgery occurred more often in participants with tier 1 than those with non-tier 1 variants. CONCLUSION Relevant phenotypic traits were observed in 57% whereas a clinical outcome occurred in 19% of participants with actionable genomic variants in the year following RoR.
Collapse
|
13
|
Allen CG, Peterson S, Khoury MJ, Brody LC, McBride CM. A scoping review of social and behavioral science research to translate genomic discoveries into population health impact. Transl Behav Med 2021; 11:901-911. [PMID: 32902617 PMCID: PMC8240657 DOI: 10.1093/tbm/ibaa076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since the completion of the Human Genome Project, progress toward translating genomic research discoveries to address population health issues has been limited. Several meetings of social and behavioral scientists have outlined priority research areas where advancement of translational research could increase population health benefits of genomic discoveries. In this review, we track the pace of progress, study size and design, and focus of genomics translational research from 2012 to 2018 and its concordance with five social and behavioral science recommended priorities. We conducted a review of the literature following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Guidelines for Scoping Reviews. Steps involved completing a search in five databases and a hand search of bibliographies of relevant literature. Our search (from 2012 to 2018) yielded 4,538 unique studies; 117 were included in the final analyses. Two coders extracted data including items from the PICOTS framework. Analysis included descriptive statistics to help identify trends in pace, study size and design, and translational priority area. Among the 117 studies included in our final sample, nearly half focused on genomics applications that have evidence to support translation or implementation into practice (Centers for Disease Control and Prevention Tier 1 applications). Common study designs were cross-sectional (40.2%) and qualitative (24.8%), with average sample sizes of 716 across all studies. Most often, studies addressed public understanding of genetics and genomics (33.3%), risk communication (29.1%), and intervention development and testing of interventions to promote behavior change (19.7%). The number of studies that address social and behavioral science priority areas is extremely limited and the pace of this research continues to lag behind basic science advances. Much of the research identified in this review is descriptive and related to public understanding, risk communication, and intervention development and testing of interventions to promote behavior change. The field has been slow to develop and evaluate public health-friendly interventions and test implementation approaches that could enable health benefits and equitable access to genomic discoveries. As the completion of the human genome approaches its 20th anniversary, full engagement of transdisciplinary efforts to address translation challenges will be required to close this gap.
Collapse
Affiliation(s)
- Caitlin G Allen
- Behavioral, Social and Health Education Sciences Department, Emory University, Atlanta, GA, USA
| | - Shenita Peterson
- Woodruff Health Science Center Library, Emory University, Atlanta, GA, USA
| | - Muin J Khoury
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lawrence C Brody
- Gene and Environment Interaction Section, National Human Genome Research Institute, Bethesda, MD, USA
| | - Colleen M McBride
- Behavioral, Social and Health Education Sciences Department, Emory University, Atlanta, GA, USA
| |
Collapse
|
14
|
Khoury MJ, Dotson WD. From genes to public health: are we ready for DNA-based population screening? Genet Med 2021; 23:996-998. [PMID: 33790422 DOI: 10.1038/s41436-021-01141-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Affiliation(s)
- Muin J Khoury
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - W David Dotson
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
15
|
Peshkin BN, Ladd MK, Isaacs C, Segal H, Jacobs A, Taylor KL, Graves KD, O'Neill SC, Schwartz MD. The Genetic Education for Men (GEM) Trial: Development of Web-Based Education for Untested Men in BRCA1/2-Positive Families. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2021; 36:72-84. [PMID: 31402434 PMCID: PMC7010546 DOI: 10.1007/s13187-019-01599-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Cascade testing for hereditary breast/ovarian cancer is an important public health priority. Increasing attention has been paid to the relevance of testing for men within BRCA1/2-positive families given that such testing may provide important information about their cancer risks, particularly for prostate cancer, and risks to their offspring. However, men are much less likely to seek genetic counseling and testing than their at-risk female relatives. To facilitate access to pre-test information and testing, we developed a web-based intervention (WI) for men that we are evaluating in a pilot randomized controlled trial (RCT). This paper describes three phases of research in the development of the WI: (1) formative (qualitative) research among men from BRCA1/2 families to assess needs and preferences for education; (2) a detailed description of the organization, format, and content of the WI; and (3) usability testing. We discuss the aims and hypotheses of the pilot RCT in which the WI is being compared with an enhanced usual care condition among at-risk men. We expect that the WI described here will foster informed decisions and lead to increased use of BRCA1/2 counseling and testing, potentially yielding improved cancer control outcomes for this understudied group, and for their at-risk relatives.
Collapse
Affiliation(s)
- Beth N Peshkin
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA.
| | - Mary Kate Ladd
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Claudine Isaacs
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Hannah Segal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Aryana Jacobs
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Kathryn L Taylor
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Kristi D Graves
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Suzanne C O'Neill
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| | - Marc D Schwartz
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
- Jess and Mildred Fisher Center for Hereditary Cancer and Clinical Genomics Research, Georgetown University, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC, 20007, USA
| |
Collapse
|
16
|
Allen CG, Green RF, Bowen S, Dotson WD, Yu W, Khoury MJ. Challenges and Opportunities for Communication about the Role of Genomics in Public Health. Public Health Genomics 2021; 24:67-74. [PMID: 33445172 DOI: 10.1159/000512485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/22/2020] [Indexed: 11/19/2022] Open
Abstract
Despite growing awareness about the potential for genomic information to improve population health, lingering communication challenges remain in describing the role of genomics in public health programs. Identifying and addressing these challenges provide an important opportunity for appropriate communication to ensure the translation of genomic discoveries for public health benefits. In this commentary, we describe 5 common communication challenges encountered by the Centers for Disease Control and Prevention's Office of Genomics and Precision Public Health based on over 20 years of experience in the field. These include (1) communicating that using genomics to assess rare diseases can have an impact on public health; (2) providing evidence that genetic factors can add important information to environmental, behavioral, and social determinants of health; (3) communicating that although genetic factors are nonmodifiable, they can increase the impact of public health programs and communication strategies; (4) addressing the concern that genomics is not ready for clinical practice; and (5) communicating that genomics is valuable beyond the domain of health care and can be integrated as part of public health programs. We discuss opportunities for addressing these communication challenges and provide examples of ongoing approaches to communication about the role of genomics in public health to the public, researchers, and practitioners.
Collapse
Affiliation(s)
- Caitlin G Allen
- Department of Behavioral, Social, and Health Education Sciences, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Ridgely Fisk Green
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Scott Bowen
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - W David Dotson
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wei Yu
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Muin J Khoury
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, Georgia, USA,
| |
Collapse
|
17
|
Strategic vision for improving human health at The Forefront of Genomics. Nature 2020; 586:683-692. [PMID: 33116284 DOI: 10.1038/s41586-020-2817-4] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Starting with the launch of the Human Genome Project three decades ago, and continuing after its completion in 2003, genomics has progressively come to have a central and catalytic role in basic and translational research. In addition, studies increasingly demonstrate how genomic information can be effectively used in clinical care. In the future, the anticipated advances in technology development, biological insights, and clinical applications (among others) will lead to more widespread integration of genomics into almost all areas of biomedical research, the adoption of genomics into mainstream medical and public-health practices, and an increasing relevance of genomics for everyday life. On behalf of the research community, the National Human Genome Research Institute recently completed a multi-year process of strategic engagement to identify future research priorities and opportunities in human genomics, with an emphasis on health applications. Here we describe the highest-priority elements envisioned for the cutting-edge of human genomics going forward-that is, at 'The Forefront of Genomics'.
Collapse
|
18
|
Deverka PA, Douglas MP, Phillips KA. Use of Real-World Evidence in US Payer Coverage Decision-Making for Next-Generation Sequencing-Based Tests: Challenges, Opportunities, and Potential Solutions. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:540-550. [PMID: 32389218 PMCID: PMC7219085 DOI: 10.1016/j.jval.2020.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/26/2020] [Accepted: 02/02/2020] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Given the potential of real-world evidence (RWE) to inform understanding of the risk-benefit profile of next-generation sequencing (NGS)-based testing, we undertook a study to describe the current landscape of whether and how payers use RWE as part of their coverage decision making and potential solutions for overcoming barriers. METHODS We performed a scoping literature review of existing RWE evidentiary frameworks for evaluating new technologies and identified barriers to clinical integration and evidence gaps for NGS. We synthesized findings as potential solutions for improving the relevance and utility of RWE for payer decision-making. RESULTS Payers require evidence of clinical utility to inform coverage decisions, yet we found a relatively small number of published RWE studies, and these are predominately focused on oncology, pharmacogenomics, and perinatal/pediatric testing. We identified 3 categories of innovation that may help address the current undersupply of RWE studies for NGS: (1) increasing use of RWE to inform outcomes-based contracting for new technologies, (2) precision medicine initiatives that integrate clinical and genomic data and enable data sharing, and (3) Food and Drug Administration reforms to encourage the use of RWE. Potential solutions include development of data and evidence review standards, payer engagement in RWE study design, use of incentives and partnerships to lower the barriers to RWE generation, education of payers and providers concerning the use of RWE and NGS, and frameworks for conducting outcomes-based contracting for NGS. CONCLUSIONS We provide numerous suggestions to overcome the data, methodologic, infrastructure, and policy challenges constraining greater integration of RWE in assessments of NGS.
Collapse
Affiliation(s)
| | - Michael P Douglas
- Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California at San Francisco, San Francisco, CA, USA
| | - Kathryn A Phillips
- Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California at San Francisco, San Francisco, CA, USA; Philip R. Lee Institute for Health Policy, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
19
|
Abstract
Purpose of review: Identification of Lynch syndrome is important from an individual patient and public health standpoint. As paradigms for Lynch syndrome diagnosis have shifted in recent years, this review will discuss rationale and limitations for current strategies as well as provide an overview of future directions in the field. Recent findings: In recent years, the use of clinical criteria and risk scores for identification of Lynch syndrome have been augmented by universal testing of all newly diagnosed colorectal cancers with molecular methods to screen for mismatch repair deficiency with high sensitivity and specificity. Studies of implementation and outcomes of universal testing in clinical practice have demonstrated significant heterogeneity that results in suboptimal uptake and contributes to disparities in diagnosis. Emerging technologies, such as next-generation sequencing, hold significant promise as a screening strategy for Lynch syndrome. Summary: Universal testing for Lynch syndrome is being performed with increasing frequency, although real-world outcomes have demonstrated room for improvement. Future directions in Lynch syndrome diagnosis will involve optimization of universal testing workflow and application of new genetics technologies.
Collapse
|
20
|
Tracking human genes along the translational continuum. NPJ Genom Med 2019; 4:25. [PMID: 31632691 PMCID: PMC6795796 DOI: 10.1038/s41525-019-0100-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/03/2019] [Indexed: 12/03/2022] Open
Abstract
Understanding the drivers of research on human genes is a critical component to success of translation efforts of genomics into medicine and public health. Using publicly available curated online databases we sought to identify specific genes that are featured in translational genetic research in comparison to all genomics research publications. Articles in the CDC’s Public Health Genomics and Precision Health Knowledge Base were stratified into studies that have moved beyond basic research to population and clinical epidemiologic studies (T1: clinical and population human genome epidemiology research), and studies that evaluate, implement, and assess impact of genes in clinical and public health areas (T2+: beyond bench to bedside). We examined gene counts and numbers of publications within these phases of translation in comparison to all genes from Medline. We are able to highlight those genes that are moving from basic research to clinical and public health translational research, namely in cancer and a few genetic diseases with high penetrance and clinical actionability. Identifying human genes of translational value is an important step towards determining an evidence-based trajectory of the human genome in clinical and public health practice over time.
Collapse
|
21
|
Bush WS, Cooke Bailey JN, Beno MF, Crawford DC. Bridging the Gaps in Personalized Medicine Value Assessment: A Review of the Need for Outcome Metrics across Stakeholders and Scientific Disciplines. Public Health Genomics 2019; 22:16-24. [PMID: 31454805 PMCID: PMC6752968 DOI: 10.1159/000501974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/07/2019] [Indexed: 12/14/2022] Open
Abstract
Despite monumental advances in genomics, relatively few health care provider organizations in the United States offer personalized or precision medicine as part of the routine clinical workflow. The gaps between research and applied genomic medicine may be a result of a cultural gap across various stakeholders representing scientists, clinicians, patients, policy makers, and third party payers. Scientists are trained to assess the health care value of genomics by either quantifying population-scale effects, or through the narrow lens of clinical trials where the standard of care is compared with the predictive power of a single or handful of genetic variants. While these metrics are an essential first step in assessing and documenting the clinical utility of genomics, they are rarely followed up with other assessments of health care value that are critical to stakeholders who use different measures to define value. The limited value assessment in both the research and implementation science of precision medicine is likely due to necessary logistical constraints of these teams; engaging bioethicists, health care economists, and individual patient belief systems is incredibly daunting for geneticists and informaticians conducting research. In this narrative review, we concisely describe several definitions of value through various stakeholder viewpoints. We highlight the existing gaps that prevent clinical translation of scientific findings generally as well as more specifically using two present-day, extreme scenarios: (1) genetically guided warfarin dosing representing a handful of genetic markers and more than 10 years of basic and translational research, and (2) next-generation sequencing representing genome-dense data lacking substantial evidence for implementation. These contemporary scenarios highlight the need for various stakeholders to broadly adopt frameworks designed to define and collect multiple value measures across different disciplines to ultimately impact more universal acceptance of and reimbursement for genomic medicine.
Collapse
Affiliation(s)
- William S Bush
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jessica N Cooke Bailey
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark F Beno
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana C Crawford
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA,
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA,
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA,
| |
Collapse
|
22
|
|
23
|
Ponte A, Greenberg S, Greendale K, Senier L. Moving the Needle on Action Around Evidence-Based Screening for Hereditary Conditions: Preparing State Chronic Disease Directors to Advance Precision Public Health. Public Health Rep 2019; 134:228-233. [PMID: 30897048 PMCID: PMC6505330 DOI: 10.1177/0033354919834588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Amy Ponte
- College of Health Sciences, Walden University, Minneapolis, MN, USA
| | | | - Karen Greendale
- Department of Health Policy, Management and Behavior, University at Albany School of Public Health, Albany, NY, USA
| | - Laura Senier
- Department of Sociology & Anthropology and Department of Health Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
24
|
Affiliation(s)
- Charles Auffray
- European Institute for Systems Biology and Medicine (EISBM), Vourles, France.
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge, CB2 1GA, UK
- Computational and Systems Medicine, Department of Surgery and Oncology, Imperial College London, London, SW7 2AZ, UK
| | - Muin J Khoury
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GA, 30329, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Baylor Plaza, Houston, TX, 77030, USA
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstraße, 70376, Stuttgart, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle, 72076, Tübingen, Germany
| |
Collapse
|
25
|
Vassy JL, Stone A, Callaghan JT, Mendes M, Meyer LJ, Pratt VM, Przygodzki RM, Scheuner MT, Wang-Rodriguez J, Schichman SA. Pharmacogenetic testing in the Veterans Health Administration (VHA): policy recommendations from the VHA Clinical Pharmacogenetics Subcommittee. Genet Med 2019; 21:382-390. [PMID: 29858578 PMCID: PMC6274593 DOI: 10.1038/s41436-018-0057-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/26/2018] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The Veterans Health Administration (VHA) Clinical Pharmacogenetics Subcommittee is charged with making recommendations about whether specific pharmacogenetic tests should be used in healthcare at VHA facilities. We describe a process to inform VHA pharmacogenetic testing policy. METHODS After developing consensus definitions of clinical validity and utility, the Subcommittee identified salient drug-gene pairs with potential clinical application in VHA. Members met monthly to discuss each drug-gene pair, the evidence of clinical utility for the associated pharmacogenetic test, and any VHA-specific testing considerations. The Subcommittee classified each test as strongly recommended, recommended, or not routinely recommended before drug initiation. RESULTS Of 30 drug-gene pair tests reviewed, the Subcommittee classified 4 (13%) as strongly recommended, including HLA-B*15:02 for carbamazepine-associated Stevens-Johnston syndrome and G6PD for rasburicase-associated hemolytic anemia; 12 (40%) as recommended, including CYP2D6 for codeine toxicity; and 14 (47%) as not routinely recommended, such as CYP2C19 for clopidogrel dosing. CONCLUSION Only half of drug-gene pairs with high clinical validity received Subcommittee support for policy promoting their widespread use across VHA. The Subcommittee generally found insufficient evidence of clinical utility or available, effective alternative strategies for the remainders. Continual evidence review and rigorous outcomes research will help promote the translation of pharmacogenetic discovery to healthcare.
Collapse
Affiliation(s)
- Jason L Vassy
- Section of General Internal Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - John T Callaghan
- Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Laurence J Meyer
- Office of Specialty Care Services, Veterans Health Administration, Washington, DC, USA
| | - Victoria M Pratt
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ronald M Przygodzki
- Office of Research & Development, US Department of Veterans Affairs, Washington, DC, USA
| | - Maren T Scheuner
- Division of Medical Genetics, Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Jessica Wang-Rodriguez
- VA San Diego Healthcare System, San Diego, California, USA
- University of California San Diego, San Diego, California, USA
| | - Steven A Schichman
- Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
26
|
Duquette D. Implementation of public health genomics and applications to public health dentistry. J Public Health Dent 2019; 80 Suppl 1:S37-S42. [PMID: 30687926 DOI: 10.1111/jphd.12307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/18/2018] [Accepted: 12/28/2018] [Indexed: 11/30/2022]
Abstract
National and state public health genomics efforts exist to effectively and responsibly translate genome-based knowledge to improve population health and reduce health disparities. Over the past two decades, public health genomics efforts have utilized the core public health functions of assessment, policy development, and assurance. Current evidence for a small number of genomic applications suggests that many lives could be saved if these were implemented in recommended populations. With the drastic increase in new genetic tests and technologies, multidisciplinary public health genomics efforts that should include public health dentistry are of greater importance. There is a need to integrate public health dentistry in efforts to increase use of evidence-based genomic tests and services to improve health outcomes. Additionally, public health genomic efforts also are utilized to promote awareness about the insufficient evidence of the validity, utility and ethical, legal, and social implications for the vast majority of genomic tests. This is demonstrated by a recent genetic testing policy statement and educational resources from the American Dental Association. These organizational efforts should be considered in other realms of public health genomics to ensure that only genetic tests and preventive services with sufficient evidence for use are being implemented in clinical and public health.
Collapse
Affiliation(s)
- Debra Duquette
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
27
|
Green RF, Ari M, Kolor K, Dotson WD, Bowen S, Habarta N, Rodriguez JL, Richardson LC, Khoury MJ. Evaluating the role of public health in implementation of genomics-related recommendations: a case study of hereditary cancers using the CDC Science Impact Framework. Genet Med 2019; 21:28-37. [PMID: 29907802 PMCID: PMC6295277 DOI: 10.1038/s41436-018-0028-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/20/2018] [Indexed: 01/21/2023] Open
Abstract
Public health plays an important role in ensuring access to interventions that can prevent disease, including the implementation of evidence-based genomic recommendations. We used the Centers for Disease Control and Prevention (CDC) Science Impact Framework to trace the impact of public health activities and partnerships on the implementation of the 2009 Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Lynch Syndrome screening recommendation and the 2005 and 2013 United States Preventive Services Task Force (USPSTF) BRCA1 and BRCA2 testing recommendations.The EGAPP and USPSTF recommendations have each been cited by >300 peer-reviewed publications. CDC funds selected states to build capacity to integrate these recommendations into public health programs, through education, policy, surveillance, and partnerships. Most state cancer control plans include genomics-related goals, objectives, or strategies. Since the EGAPP recommendation, major public and private payers now provide coverage for Lynch Syndrome screening for all newly diagnosed colorectal cancers. National guidelines and initiatives, including Healthy People 2020, included similar recommendations and cited the EGAPP and USPSTF recommendations. However, disparities in implementation based on race, ethnicity, and rural residence remain challenges. Public health achievements in promoting the evidence-based use of genomics for the prevention of hereditary cancers can inform future applications of genomics in public health.
Collapse
Affiliation(s)
- Ridgely Fisk Green
- Carter Consulting and Office of Public Health Genomics, Division of Public Health Information Dissemination, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
| | - Mary Ari
- Office of the Director, Office of the Associate Director for Science, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katherine Kolor
- Office of Public Health Genomics, Division of Public Health Information Dissemination, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - W David Dotson
- Office of Public Health Genomics, Division of Public Health Information Dissemination, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Scott Bowen
- Office of Public Health Genomics, Division of Public Health Information Dissemination, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nancy Habarta
- Division of Public Health Information Dissemination, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Juan L Rodriguez
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Lisa C Richardson
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Muin J Khoury
- Office of Public Health Genomics, Division of Public Health Information Dissemination, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Sun YV, Damrauer SM, Hui Q, Assimes TL, Ho YL, Natarajan P, Klarin D, Huang J, Lynch J, DuVall SL, Pyarajan S, Honerlaw JP, Gaziano JM, Cho K, Rader DJ, O’Donnell CJ, Tsao PS, Wilson PWF. Effects of Genetic Variants Associated with Familial Hypercholesterolemia on Low-Density Lipoprotein-Cholesterol Levels and Cardiovascular Outcomes in the Million Veteran Program. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e002192. [PMID: 31106297 PMCID: PMC6516478 DOI: 10.1161/circgen.118.002192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background Familial hypercholesterolemia (FH) is characterized by inherited high levels of low-density lipoprotein cholesterol (LDL-C) and premature coronary heart disease (CHD). Over a thousand low-frequency variants in LDLR, APOB and PCSK9 have been implicated in FH but few have been examined at the population level. We aim to estimate the phenotypic effects of a subset of FH variants on LDL-C and clinical outcomes among 331,107 multi-ethnic participants. Methods We examined the individual and collective association between putatively pathogenic FH variants included on the MVP biobank array and the maximum LDL-C level over an interval of 15 years (maxLDL). We assessed the collective effect on clinical outcomes by leveraging data from 61.7 million clinical encounters. Results We found 8 out of 16 putatively pathogenic FH variants with ≥30 observed carriers to be significantly associated with elevated maxLDL (9.4-80.2 mg/dL). Phenotypic effects were similar for European and African Americans despite substantial differences in carrier frequencies. Based on observed effects on maxLDL, we identified a total of 748 carriers (1:443) who had elevated maxLDL (36.5±1.4 mg/dL, p=1.2×10-152), and higher prevalence of clinical diagnoses related to hypercholesterolemia and CHD in a phenome-wide scan. Adjusted for maxLDL, FH variants collectively associated with higher prevalence of CHD (odds ratio, 1.59 [95% CI 1.36-1.86], p=1.1×10-8) but not peripheral artery disease. Conclusions The distribution and phenotypic effects of putatively pathogenic FH variants were heterogeneous within and across variants. More robust evidence of genotype-phenotype associations of FH variants in multi-ethnic populations is needed to accurately infer at-risk individuals from genetic screening.
Collapse
Affiliation(s)
- Yan V. Sun
- Department of Epidemiology, Emory University Rollins School
of Public Health
- Department of Biomedical Informatics, Emory University
School of Medicine, Atlanta, GA
| | - Scott M. Damrauer
- Corporal Michael Crescenz VA Medical Center,University of
Pennsylvania, Philadelphia, PA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School
of Public Health
| | - Themistocles L. Assimes
- VA Palo Alto Health Care System, Department of Medicine,
Stanford University School of Medicine, Stanford, CA
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research
Center, Massachusetts General Hospital, Boston, MA. Department of Medicine, Harvard
Medical School, Program in Medical & Population Genetics, Broad Institute of
Harvard & MIT, Cambridge
| | - Derek Klarin
- Massachusetts General Hospital, Boston, MA, Broad Institute
of Harvard & MIT, Cambridge
| | - Jie Huang
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
| | - Julie Lynch
- University of Massachusetts College of Nursing & Health
Sciences, Boston, MA
- Department of Veterans Affairs Salt Lake City Health Care
System
| | - Scott L. DuVall
- Department of Veterans Affairs Salt Lake City Health Care
System
- University of Utah, School of Medicine, Salt Lake City,
UT
| | - Saiju Pyarajan
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
| | - Jacqueline P. Honerlaw
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
| | - J. Michael Gaziano
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
- Department of Medicine, Brigham and Women’s
Hospital, Boston, MA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
- Department of Medicine, Brigham and Women’s
Hospital, Boston, MA
| | - Daniel J. Rader
- Perlman School of Medicine, University of Pennsylvania,
Philadelphia, PA
| | - Christopher J. O’Donnell
- Massachusetts Veterans Epidemiology Research and
Information Center (MAVERIC), VA Boston Healthcare System, Boston
- Harvard Medical School, Boston, MA
| | - Philip S. Tsao
- VA Palo Alto Health Care System, Department of Medicine,
Stanford University School of Medicine, Stanford, CA
| | - Peter W. F. Wilson
- Atlanta VA Medical Center and Emory Clinical
Cardiovascular Research Institute, Atlanta, GA
| |
Collapse
|
29
|
Khoury MJ, Feero WG, Chambers DA, Brody LE, Aziz N, Green RC, Janssens ACJ, Murray MF, Rodriguez LL, Rutter JL, Schully SD, Winn DM, Mensah GA. A collaborative translational research framework for evaluating and implementing the appropriate use of human genome sequencing to improve health. PLoS Med 2018; 15:e1002631. [PMID: 30071015 PMCID: PMC6071954 DOI: 10.1371/journal.pmed.1002631] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In a Policy Forum, Muin Khoury and colleagues discuss research on the clinical application of genome sequencing data.
Collapse
Affiliation(s)
- Muin J. Khoury
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| | - W. Gregory Feero
- Maine-Dartmouth Family Medicine Residency Program, Augusta, Maine, United States of America
| | - David A. Chambers
- Division of Cancer Control and Population Sciences, National Cancer Institute, NIH, Rockville, Maryland, United States of America
| | - Lawrence E. Brody
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nazneen Aziz
- Kaiser Permanente, Oakland, California, United States of America
| | - Robert C. Green
- Brigham and Women’s Hospital, Broad Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - A. Cecile J.W. Janssens
- Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Michael F. Murray
- Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Laura Lyman Rodriguez
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joni L. Rutter
- All of Us Research Program, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sheri D. Schully
- Office of Disease Prevention, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Deborah M. Winn
- Division of Cancer Control and Population Sciences, National Cancer Institute, NIH, Rockville, Maryland, United States of America
| | - George A. Mensah
- Center for Translation Research and Implementation Science, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| |
Collapse
|
30
|
Insurance Coverage Policies for Pharmacogenomic and Multi-Gene Testing for Cancer. J Pers Med 2018; 8:jpm8020019. [PMID: 29772692 PMCID: PMC6023380 DOI: 10.3390/jpm8020019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/17/2022] Open
Abstract
Insurance coverage policies are a major determinant of patient access to genomic tests. The objective of this study was to examine differences in coverage policies for guideline-recommended pharmacogenomic tests that inform cancer treatment. We analyzed coverage policies from eight Medicare contractors and 10 private payers for 23 biomarkers (e.g., HER2 and EGFR) and multi-gene tests. We extracted policy coverage and criteria, prior authorization requirements, and an evidence basis for coverage. We reviewed professional society guidelines and their recommendations for use of pharmacogenomic tests. Coverage for KRAS, EGFR, and BRAF tests were common across Medicare contractors and private payers, but few policies covered PML/RARA, CD25, or G6PD. Twelve payers cover at least one multi-gene test for nonsmall cell lung cancer, citing emerging clinical recommendations. Coverage policies for single and multi-gene tests for cancer treatments are relatively consistent among Medicare contractors despite the lack of national coverage determinations. In contrast, coverage for these tests varied across private payers. Patient access to tests is governed by prior authorization among eight private payers. Substantial variations in how payers address guideline-recommended pharmacogenomic tests and the common use of prior authorization underscore the need for additional studies of the effects of coverage variation on cancer care and patient outcomes.
Collapse
|
31
|
Boca SM, Panagiotou OA, Rao S, McGarvey PB, Madhavan S. Future of Evidence Synthesis in Precision Oncology: Between Systematic Reviews and Biocuration. JCO Precis Oncol 2018; 2:PO.17.00175. [PMID: 31930186 PMCID: PMC6953752 DOI: 10.1200/po.17.00175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Simina M. Boca
- Simina M. Boca, Shruti Rao, Peter B. McGarvey, and Subha Madhavan, Georgetown University Medical Center, Washington, DC; and Orestis A. Panagiotou, Brown University School of Public Health, Providence, RI
| | - Orestis A. Panagiotou
- Simina M. Boca, Shruti Rao, Peter B. McGarvey, and Subha Madhavan, Georgetown University Medical Center, Washington, DC; and Orestis A. Panagiotou, Brown University School of Public Health, Providence, RI
| | - Shruti Rao
- Simina M. Boca, Shruti Rao, Peter B. McGarvey, and Subha Madhavan, Georgetown University Medical Center, Washington, DC; and Orestis A. Panagiotou, Brown University School of Public Health, Providence, RI
| | - Peter B. McGarvey
- Simina M. Boca, Shruti Rao, Peter B. McGarvey, and Subha Madhavan, Georgetown University Medical Center, Washington, DC; and Orestis A. Panagiotou, Brown University School of Public Health, Providence, RI
| | - Subha Madhavan
- Simina M. Boca, Shruti Rao, Peter B. McGarvey, and Subha Madhavan, Georgetown University Medical Center, Washington, DC; and Orestis A. Panagiotou, Brown University School of Public Health, Providence, RI
| |
Collapse
|
32
|
Which Lynch syndrome screening programs could be implemented in the "real world"? A systematic review of economic evaluations. Genet Med 2018; 20:1131-1144. [PMID: 29300371 PMCID: PMC8660650 DOI: 10.1038/gim.2017.244] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
Abstract
Purpose Lynch syndrome (LS) screening can significantly reduce cancer morbidity and mortality in mutation carriers. Our aim was to identify cost-effective LS screening programs that can be implemented in the “real world.” Methods We performed a systematic review of full economic evaluations of genetic screening for LS in different target populations; health outcomes were estimated in life-years gained or quality-adjusted life-years. Results Overall, 20 studies were included in the systematic review. Based on the study populations, we identified six categories of LS screening program: colorectal cancer (CRC)–based, endometrial cancer–based, general population–based, LS family registry–based, cascade testing–based, and genetics clinic–based screening programs. We performed an in-depth analysis of CRC-based LS programs, classifying them into three additional subcategories: universal, age-targeted, and selective. In five studies, universal programs based on immunohistochemistry, either alone or in combination with the BRAF test, were cost-effective compared with no screening, while in two studies age-targeted programs with a cutoff of 70 years were cost-effective when compared with age-targeted programs with lower age thresholds. Conclusion Universal or <70 years–age-targeted CRC-based LS screening programs are cost-effective and should be implemented in the “real world.”
Collapse
|
33
|
Khoury MJ, Bowen MS, Clyne M, Dotson WD, Gwinn ML, Green RF, Kolor K, Rodriguez JL, Wulf A, Yu W. From public health genomics to precision public health: a 20-year journey. Genet Med 2017; 20:574-582. [PMID: 29240076 DOI: 10.1038/gim.2017.211] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/20/2017] [Indexed: 12/20/2022] Open
Abstract
In this paper, we review the evolution of the field of public health genomics in the United States in the past two decades. Public health genomics focuses on effective and responsible translation of genomic science into population health benefits. We discuss the relationship of the field to the core public health functions and essential services, review its evidentiary foundation, and provide examples of current US public health priorities and applications. We cite examples of publications to illustrate how Genetics in Medicine reflected the evolution of the field. We also reflect on how public-health genomics is contributing to the emergence of "precision public health" with near-term opportunities offered by the US Precision Medicine (AllofUs) Initiative.
Collapse
Affiliation(s)
- Muin J Khoury
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - M Scott Bowen
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mindy Clyne
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland, USA
| | - W David Dotson
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Marta L Gwinn
- Office of Advanced Molecular Detection, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ridgely Fisk Green
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katherine Kolor
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Juan L Rodriguez
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Anja Wulf
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wei Yu
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
34
|
Wu AC, Mazor KM, Ceccarelli R, Loomer S, Lu CY. Access to Guideline-Recommended Pharmacogenomic Tests for Cancer Treatments: Experience of Providers and Patients. J Pers Med 2017; 7:jpm7040017. [PMID: 29140263 PMCID: PMC5748629 DOI: 10.3390/jpm7040017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/28/2017] [Accepted: 11/08/2017] [Indexed: 01/20/2023] Open
Abstract
Genomic tests are the fastest growing sector in medicine and medical science, yet there remains a dearth of research on access to pharmacogenomic tests and medications. The objective of this study is to explore providers' and patients' experiences and views on test access as well as strategies used for gaining access. We interviewed clinicians who prescribed medications that should be guided by pharmacogenomic testing and patients who received those prescriptions. We organized the themes into the four dimensions suggested by the World Health Organization framework on access to medications and health technologies. Guideline-recommended pharmacogenomic tests for cancer care are generally available, although the timeliness of return of test results is sometimes suboptimal. Accessibility of pharmacogenomic tests is made challenging by the process of ordering pharmacogenomic tests, which is time-consuming. Affordability is a barrier to some patients as expressed by both providers and patients, who noted that the cost of pharmacogenomic tests and medications is high. Acceptability of the tests is high as both providers and patients view the tests positively. Understanding challenges to accessing pharmacogenomic tests will allow policymakers to develop policies that streamline access to genomics-based technologies to improve population health.
Collapse
Affiliation(s)
- Ann Chen Wu
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, 401 Park Drive, Suite 401, Boston, MA 02215, USA.
| | - Kathleen M Mazor
- Meyers Primary Care Institute, A Joint Endeavor of the University of Massachusetts Medical School, Reliant Medical Group and Fallon Health; 630 Plantation Street, Worcester, MA 02215, USA.
| | - Rachel Ceccarelli
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, 401 Park Drive, Suite 401, Boston, MA 02215, USA.
| | - Stephanie Loomer
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, 401 Park Drive, Suite 401, Boston, MA 02215, USA.
| | - Christine Y Lu
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, 401 Park Drive, Suite 401, Boston, MA 02215, USA.
| |
Collapse
|
35
|
Meagher KM, McGowan ML, Settersten RA, Fishman JR, Juengst ET. Precisely Where Are We Going? Charting the New Terrain of Precision Prevention. Annu Rev Genomics Hum Genet 2017; 18:369-387. [PMID: 28441061 PMCID: PMC6203331 DOI: 10.1146/annurev-genom-091416-035222] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In addition to genetic data, precision medicine research gathers information about three factors that modulate gene expression: lifestyles, environments, and communities. The relevant research tools-epidemiology, environmental assessment, and socioeconomic analysis-are those of public health sciences rather than molecular biology. Because these methods are designed to support inferences and interventions addressing population health, the aspirations of this research are expanding from individualized treatment toward precision prevention in public health. The purpose of this review is to explore the emerging goals and challenges of such a shift to help ensure that the genomics community and public policy makers understand the ethical issues at stake in embracing and pursuing precision prevention. Two emerging goals bear special attention in this regard: (a) public health risk reduction strategies, such as screening, and (b) the application of genomic variation studies to understand and reduce health disparities among population groups.
Collapse
Affiliation(s)
- Karen M Meagher
- Center for Genomics and Society, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599;
| | - Michelle L McGowan
- Ethics Center, Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229;
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio 45229
- Department of Women's, Gender, and Sexuality Studies, University of Cincinnati, Cincinnati, Ohio 45221
| | - Richard A Settersten
- Human Development and Family Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon 97331;
| | - Jennifer R Fishman
- Biomedical Ethics Unit, Department of Social Studies of Medicine, McGill University, Montreal, Quebec H3A 1X1, Canada;
| | - Eric T Juengst
- Center for Bioethics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599;
| |
Collapse
|
36
|
A proposed approach to accelerate evidence generation for genomic-based technologies in the context of a learning health system. Genet Med 2017; 20:390-396. [PMID: 28796238 DOI: 10.1038/gim.2017.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
Genomic technologies should demonstrate analytical and clinical validity and clinical utility prior to wider adoption in clinical practice. However, the question of clinical utility remains unanswered for many genomic technologies. In this paper, we propose three building blocks for rapid generation of evidence on clinical utility of promising genomic technologies that underpin clinical and policy decisions. We define promising genomic tests as those that have proven analytical and clinical validity. First, risk-sharing agreements could be implemented between payers and manufacturers to enable temporary coverage that would help incorporate promising technologies into routine clinical care. Second, existing data networks, such as the Sentinel Initiative and the National Patient-Centered Clinical Research Network (PCORnet) could be leveraged, augmented with genomic information to track the use of genomic technologies and monitor clinical outcomes in millions of people. Third, endorsement and engagement from key stakeholders will be needed to establish this collaborative model for rapid evidence generation; all stakeholders will benefit from better information regarding the clinical utility of these technologies. This collaborative model can create a multipurpose and reusable national resource that generates knowledge from data gathered as part of routine care to drive evidence-based clinical practice and health system changes.
Collapse
|
37
|
Harada S, Zhou Y, Duncan S, Armstead AR, Coshatt GM, Dillon C, Brott BC, Willig J, Alsip JA, Hillegass WB, Limdi NA. Precision Medicine at the University of Alabama at Birmingham: Laying the Foundational Processes Through Implementation of Genotype-Guided Antiplatelet Therapy. Clin Pharmacol Ther 2017; 102:493-501. [PMID: 28124392 DOI: 10.1002/cpt.631] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/04/2017] [Accepted: 01/15/2017] [Indexed: 12/14/2022]
Abstract
Precision medicine entails tailoring treatment based on patients' unique characteristics. As drug therapy constitutes the cornerstone of treatment for most chronic diseases, pharmacogenomics (PGx), the study of genetic variation influencing individual response to drugs, is an important component of precision medicine. Over the past decade investigations have identified genes and single-nucleotide polymorphisms (SNPs) and quantified their effect on drug response. Parallel development of point-of-care (POC) genotyping platforms has enabled the interrogation of the genes/SNPs within a timeline conducive to the provision of care. Despite these advances, the pace of integration of genotype-guided drug therapy (GGTx) into practice has faced significant challenges. These include difficulty in identifying SNPs with sufficiently robust evidence to guide clinical decision making, lack of clinician training on how to order and use genotype data, lack of clinical decision support (CDS) to guide treatment, and limited reimbursement. The University of Alabama at Birmingham's (UAB) efforts in precision medicine were initiated to address these challenges and improve the health of the racially diverse patients we treat.
Collapse
Affiliation(s)
- S Harada
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Y Zhou
- Department of Pathology, University of Oklahoma Health Sciences Center, Norman, Oklahoma, USA
| | - S Duncan
- University of Alabama at Birmingham Health System, Birmingham, Alabama, USA
| | - A R Armstead
- University of Alabama at Birmingham Health System, Birmingham, Alabama, USA
| | - G M Coshatt
- University of Alabama at Birmingham Health System, Birmingham, Alabama, USA
| | - C Dillon
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - B C Brott
- Department of Medicine, Division of Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J Willig
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J A Alsip
- University of Alabama at Birmingham Health System, Birmingham, Alabama, USA
| | | | - N A Limdi
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
38
|
Rodriguez JL, Thomas CC, Massetti GM, Duquette D, Avner L, Iskander J, Khoury MJ, Richardson LC. CDC Grand Rounds: Family History and Genomics as Tools for Cancer Prevention and Control. MMWR-MORBIDITY AND MORTALITY WEEKLY REPORT 2016; 65:1291-1294. [DOI: 10.15585/mmwr.mm6546a3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
39
|
Yu W, Gwinn M, Dotson WD, Green RF, Clyne M, Wulf A, Bowen S, Kolor K, Khoury MJ. A knowledge base for tracking the impact of genomics on population health. Genet Med 2016; 18:1312-1314. [PMID: 27280867 DOI: 10.1038/gim.2016.63] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/06/2016] [Indexed: 01/08/2023] Open
Abstract
PURPOSE We created an online knowledge base (the Public Health Genomics Knowledge Base (PHGKB)) to provide systematically curated and updated information that bridges population-based research on genomics with clinical and public health applications. METHODS Weekly horizon scanning of a wide variety of online resources is used to retrieve relevant scientific publications, guidelines, and commentaries. After curation by domain experts, links are deposited into Web-based databases. RESULTS PHGKB currently consists of nine component databases. Users can search the entire knowledge base or search one or more component databases directly and choose options for customizing the display of their search results. CONCLUSION PHGKB offers researchers, policy makers, practitioners, and the general public a way to find information they need to understand the complicated landscape of genomics and population health.Genet Med 18 12, 1312-1314.
Collapse
Affiliation(s)
- Wei Yu
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Marta Gwinn
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.,McKing Consulting Corporation, Atlanta, Georgia, USA
| | - W David Dotson
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ridgely Fisk Green
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.,Carter Consulting, Inc., Atlanta, Georgia, USA
| | - Mindy Clyne
- Epidemiology and Genomics Research Program, National Cancer Institute, Bethesda, Maryland, USA.,Kelly Services, Troy, Michigan, USA
| | - Anja Wulf
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.,Cadence Group, Atlanta, Georgia, USA
| | - Scott Bowen
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katherine Kolor
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Muin J Khoury
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
40
|
Garzotto M, Kopp RP. Gene-expression profiling of localized prostate cancer: still miles to go before we sleep. Future Oncol 2016; 12:273-6. [DOI: 10.2217/fon.15.316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mark Garzotto
- Veterans Administration Portland Health Care System, Oregon Health & Science University, Portland, OR, USA
- Department of Urology, Oregon Health & Science University, Portland, OR, USA
- Radiation Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ryan P Kopp
- Veterans Administration Portland Health Care System, Oregon Health & Science University, Portland, OR, USA
- Department of Urology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
41
|
Robinson LS, Hendrix A, Xie XJ, Yan J, Pirzadeh-Miller S, Pritzlaff M, Read P, Pass S, Euhus D, Ross TS. Prediction of Cancer Prevention: From Mammogram Screening to Identification of BRCA1/2 Mutation Carriers in Underserved Populations. EBioMedicine 2015; 2:1827-33. [PMID: 26870808 PMCID: PMC4740331 DOI: 10.1016/j.ebiom.2015.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The US Preventative Service Task Force recommends that physicians perform a genetic risk assessment to identify women at risk for BRCA1/2 mutations associated with hereditary breast and ovarian cancer (HBOC) syndrome. However, outcomes data after a diagnosis of HBOC syndrome especially in diverse populations, are minimal. Here we asked if genetic screening of high-risk underserved women identified in the mammogram population reduces cancer incidence. METHODS We evaluated 61,924 underserved women at screening mammography for family histories suggestive of HBOC syndrome over the course of 21 months. Data were collected retrospectively from patients at two safety net hospitals through chart review. A computer model was used to calculate the long-term effect of this screening on cancer incidence by assessing both the mutation detection rate and the completion of prophylactic surgeries in BRCA1/2 mutation carriers. FINDINGS We identified 20 of the 85 (23.5%) expected BRCA1/2 mutation carriers in the underserved population. The frequencies of prophylactic mastectomies and oophorectomies in the mutation carriers were 25% and 40%, respectively. Using these data, our model predicted only an 8.8% reduction in both breast and ovarian cancer in the underserved patients. This contrasts with a 57% reduction in breast cancer and 51% reduction in ovarian cancer in an insured reference population. Our data indicate that underserved patients with HBOC syndrome are difficult to identify and when identified are limited in their ability to adhere to NCCN guidelines for cancer prevention. INTERPRETATION Screening for women at risk for HBOC syndrome in mammogram populations will only prevent cancers if we can increase compliance with management guidelines. This study provides prototypic baseline data for step-wise analysis of the efficacy of the use of family history analysis in the mammography setting for detection and management of HBOC syndrome.
Collapse
Affiliation(s)
- Linda S Robinson
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA
| | - Ashley Hendrix
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA
| | - Xian-Jin Xie
- Department of Biostatistics and Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, USA; Harold Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center,Dallas, TX, USA
| | - Jingsheng Yan
- Department of Biostatistics and Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, USA; Harold Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center,Dallas, TX, USA
| | - Sara Pirzadeh-Miller
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA
| | - Mary Pritzlaff
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA
| | - Parker Read
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA
| | - Sarah Pass
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA
| | - David Euhus
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Theodora S Ross
- Department of Cancer Genetics, University of Texas Southwestern Medical Center's Harold Simmons Comprehensive Cancer Center, Dallas and Moncrief Cancer Institute, Fort Worth, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
42
|
Fisk Green R, Dotson WD, Bowen S, Kolor K, Khoury MJ. Genomics in Public Health: Perspective from the Office of Public Health Genomics at the Centers for Disease Control and Prevention (CDC). Healthcare (Basel) 2015; 3:830-7. [PMID: 26636032 PMCID: PMC4666313 DOI: 10.3390/healthcare3030830] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/08/2015] [Indexed: 12/19/2022] Open
Abstract
The national effort to use genomic knowledge to save lives is gaining momentum, as illustrated by the inclusion of genomics in key public health initiatives, including Healthy People 2020, and the recent launch of the precision medicine initiative. The Office of Public Health Genomics (OPHG) at the Centers for Disease Control and Prevention (CDC) partners with state public health departments and others to advance the translation of genome-based discoveries into disease prevention and population health. To do this, OPHG has adopted an "identify, inform, and integrate" model: identify evidence-based genomic applications ready for implementation, inform stakeholders about these applications, and integrate these applications into public health at the local, state, and national level. This paper addresses current and future work at OPHG for integrating genomics into public health programs.
Collapse
Affiliation(s)
- Ridgely Fisk Green
- Carter Consulting, Inc. and Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - W. David Dotson
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; E-Mails: (W.D.D.); (S.B.); (K.K.); (M.J.K.)
| | - Scott Bowen
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; E-Mails: (W.D.D.); (S.B.); (K.K.); (M.J.K.)
| | - Katherine Kolor
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; E-Mails: (W.D.D.); (S.B.); (K.K.); (M.J.K.)
| | - Muin J. Khoury
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; E-Mails: (W.D.D.); (S.B.); (K.K.); (M.J.K.)
| |
Collapse
|
43
|
Khoury MJ, Evans JP. A public health perspective on a national precision medicine cohort: balancing long-term knowledge generation with early health benefit. JAMA 2015; 313:2117-8. [PMID: 26034952 PMCID: PMC4685667 DOI: 10.1001/jama.2015.3382] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Muin J Khoury
- Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - James P Evans
- Department of Genetics, University of North Carolina, Chapel Hill3Department of Medicine, University of North Carolina, Chapel Hill
| |
Collapse
|
44
|
Trosman JR, Weldon CB, Kate Kelley R, Phillips KA. Challenges of coverage policy development for next-generation tumor sequencing panels: experts and payers weigh in. J Natl Compr Canc Netw 2015; 13:311-8. [PMID: 25736008 PMCID: PMC4372087 DOI: 10.6004/jnccn.2015.0043] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Next-generation tumor sequencing (NGTS) panels, which include multiple established and novel targets across cancers, are emerging in oncology practice, but lack formal positive coverage by US payers. Lack of coverage may impact access and adoption. This study identified challenges of NGTS coverage by private payers. METHODS We conducted semi-structured interviews with 14 NGTS experts on potential NGTS benefits, and with 10 major payers, representing more than 125,000,000 enrollees, on NGTS coverage considerations. We used the framework approach of qualitative research for study design and thematic analyses and simple frequencies to further describe findings. RESULTS All interviewed payers see potential NGTS benefits, but all noted challenges to formal coverage: 80% state that inherent features of NGTS do not fit the medical necessity definition required for coverage, 70% view NGTS as a bundle of targets versus comprehensive tumor characterization and may evaluate each target individually, and 70% express skepticism regarding new evidence methods proposed for NGTS. Fifty percent of payers expressed sufficient concerns about NGTS adoption and implementation that will preclude their ability to issue positive coverage policies. CONCLUSIONS Payers perceive that NGTS holds significant promise but, in its current form, poses disruptive challenges to coverage policy frameworks. Proactive multidisciplinary efforts to define the direction for NGTS development, evidence generation, and incorporation into coverage policy are necessary to realize its promise and provide patient access. This study contributes to current literature, as possibly the first study to directly interview US payers on NGTS coverage and reimbursement.
Collapse
Affiliation(s)
- Julia R. Trosman
- UCSF Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), Department of Clinical Pharmacy, University of California, San Francisco (UCSF), San Francisco, California
- Center for Business Models in Healthcare, Chicago Illinois
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Christine B. Weldon
- UCSF Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), Department of Clinical Pharmacy, University of California, San Francisco (UCSF), San Francisco, California
- Center for Business Models in Healthcare, Chicago Illinois
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - R. Kate Kelley
- UCSF Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), Department of Clinical Pharmacy, University of California, San Francisco (UCSF), San Francisco, California
- Department of Medicine, Division of Hematology/Oncology, UCSF, San Francisco, California
| | - Kathryn A. Phillips
- UCSF Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), Department of Clinical Pharmacy, University of California, San Francisco (UCSF), San Francisco, California
- Helen Diller Family Comprehensive Cancer Center at UCSF, San Francisco, California
| |
Collapse
|
45
|
Wilson BJ, Nicholls SG. The Human Genome Project, and recent advances in personalized genomics. Risk Manag Healthc Policy 2015; 8:9-20. [PMID: 25733939 PMCID: PMC4337712 DOI: 10.2147/rmhp.s58728] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The language of “personalized medicine” and “personal genomics” has now entered the common lexicon. The idea of personalized medicine is the integration of genomic risk assessment alongside other clinical investigations. Consistent with this approach, testing is delivered by health care professionals who are not medical geneticists, and where results represent risks, as opposed to clinical diagnosis of disease, to be interpreted alongside the entirety of a patient’s health and medical data. In this review we consider the evidence concerning the application of such personalized genomics within the context of population screening, and potential implications that arise from this. We highlight two general approaches which illustrate potential uses of genomic information in screening. The first is a narrowly targeted approach in which genetic profiling is linked with standard population-based screening for diseases; the second is a broader targeting of variants associated with multiple single gene disorders, performed opportunistically on patients being investigated for unrelated conditions. In doing so we consider the organization and evaluation of tests and services, the challenge of interpretation with less targeted testing, professional confidence, barriers in practice, and education needs. We conclude by discussing several issues pertinent to health policy, namely: avoiding the conflation of genetics with biological determinism, resisting the “technological imperative”, due consideration of the organization of screening services, the need for professional education, as well as informed decision making and public understanding.
Collapse
Affiliation(s)
- Brenda J Wilson
- Department of Epidemiology and Community Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Stuart G Nicholls
- Department of Epidemiology and Community Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
46
|
Schully SD, Khoury MJ. What is translational genomics? An expanded research agenda for improving individual and population health. Appl Transl Genom 2014; 3:82-83. [PMID: 26722640 PMCID: PMC4694629 DOI: 10.1016/j.atg.2014.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
47
|
Hendershot CS. Pharmacogenetic approaches in the treatment of alcohol use disorders: addressing clinical utility and implementation thresholds. Addict Sci Clin Pract 2014; 9:20. [PMID: 25217046 PMCID: PMC4165632 DOI: 10.1186/1940-0640-9-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 09/11/2014] [Indexed: 12/22/2022] Open
Abstract
Despite advances in characterizing genetic influences on addiction liability and treatment response, clinical applications of these efforts have been slow to evolve. Although challenges to clinical translation remain, stakeholders already face decisions about evidentiary thresholds for the uptake of pharmacogenetic tests in practice. There is optimism about potential pharmacogenetic applications for the treatment of alcohol use disorders, with particular interest in the OPRM1 A118G polymorphism as a moderator of naltrexone response. Findings from human and animal studies suggest preliminary evidence for the clinical validity of this association; on this basis, arguments for clinical implementation can be made in accordance with existing frameworks for the uptake of genomic applications. However, generating evidence-based guidelines requires evaluating the clinical utility of pharmacogenetic tests. This goal will remain challenging, largely due to minimal data to inform clinical utility estimates. The pace of genomic discovery highlights the need for clinical utility and implementation research to inform future translation efforts. Near-term implementation of promising pharmacogenetic tests can help expedite this goal, generating an evidence base to enable efficient translation as additional gene-drug associations are discovered.
Collapse
Affiliation(s)
- Christian S Hendershot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| |
Collapse
|
48
|
|
49
|
Abstract
Personalized medicine is the cornerstone of medical practice. It tailors treatments for specific conditions of an affected individual. The borders of personalized medicine are defined by limitations in technology and our understanding of biology, physiology and pathology of various conditions. Current advances in technology have provided physicians with the tools to investigate the molecular makeup of the disease. Translating these molecular make-ups to actionable targets has led to the development of small molecular inhibitors. Also, detailed understanding of genetic makeup has allowed us to develop prognostic markers, better known as companion diagnostics. Current attempts in the development of drug delivery systems offer the opportunity of delivering specific inhibitors to affected cells in an attempt to reduce the unwanted side effects of drugs.
Collapse
Affiliation(s)
- Gayane Badalian-Very
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline ave, Boston, MA 02115, United States. Tel.: + 1 617 513 7940; fax: + 1 617 632 5998.
| |
Collapse
|
50
|
A horizon-prioritizing method can identify gaps among genomic application guidelines. Clin Pharmacol Ther 2014; 95:368-9. [PMID: 24646488 DOI: 10.1038/clpt.2014.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In light of the increasing need by decision makers for a method of evaluating genomic applications based on the weight of evidence for their efficacy, several agencies have developed systems of classification. Here I review the horizon-scanning method for prioritizing genomics applications as described by Dotson et al. in this issue of CPT. Using the examples of the authors' Tier 1/Green classification for KRAS and Tier 2/Yellow for TPMT, I discuss differences between the guidelines issued by the Clinical Pharmacogenetics Implementation Consortium (CPIC) and those by the National Comprehensive Cancer Network (NCCN). Additionally, I offer suggestions regarding classification of the Tier 3/Red genomics applications and the reproducibility of the data-curating algorithm of the horizon-scanning method.
Collapse
|