1
|
Wiedower J, Smith HS, Farrell CL, Parker V, Rebek L, Davis SC. Payer perspectives on genomic testing in the United States: A systematic literature review. Genet Med 2025; 27:101329. [PMID: 39556478 DOI: 10.1016/j.gim.2024.101329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
PURPOSE Health care stakeholders' perspectives on the value of genomic testing vary widely and directly affect the access and practice of genomic medicine. To our knowledge, a review of US health care payers' perspectives on genomic testing has not been performed. METHODS We conducted a systematic literature review of US payers' perspectives on genomic testing in the MEDLINE, PubMed, and Cumulative Index to Nursing and Allied Health Literature (CINAHL) databases. Of the 161 nonduplicate records screened, we summarized findings from 20 included records, and using the framework method, common domains were recorded. RESULTS Domains included clinical utility, coverage decision frameworks, potential harms, costs, paying for research, demand/pressure, the flexibility of outcomes considered, and personal utility. There was consensus on the definition of clinical utility as improved health outcomes, and the nuances of genomic testing were reported as challenging to fit within existing coverage decision frameworks. Perspectives varied on accepting broader outcomes or uses of genomic testing and whether costs influence coverage decisions. Study methodologies were heterogeneous. CONCLUSION A deeper understanding of how payers approach genomic testing may allow comparison with other stakeholders' perspectives and may identify challenges, opportunities, and solutions to align a conceptual and evidentiary framework better to demonstrate the value of genomic testing.
Collapse
Affiliation(s)
- Julie Wiedower
- Clemson University, School of Nursing, Clemson, SC; Guardant Health, Redwood City, CA.
| | - Hadley Stevens Smith
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA; Center for Bioethics, Harvard Medical School, Boston, MA
| | | | | | | | | |
Collapse
|
2
|
Liang JW, Christensen KD, Green RC, Kraft P. Evaluating the utility of multi-gene, multi-disease population-based panel testing accounting for uncertainty in penetrance estimates. NPJ Genom Med 2024; 9:30. [PMID: 38760335 PMCID: PMC11101660 DOI: 10.1038/s41525-024-00414-y] [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: 01/04/2023] [Accepted: 04/19/2024] [Indexed: 05/19/2024] Open
Abstract
Panel germline testing allows for the efficient detection of deleterious variants for multiple conditions, but the benefits and harms of identifying these variants are not always well understood. We present a multi-gene, multi-disease aggregate utility formula that allows the user to consider adding or removing each gene in a panel based on variant frequency, estimated penetrances, and subjective disutilities for testing positive but not developing the disease and testing negative but developing the disease. We provide credible intervals for utility that reflect uncertainty in penetrance estimates. Rare, highly penetrant deleterious variants tend to contribute positive net utilities for a wide variety of user-specified disutilities, even when accounting for parameter estimation uncertainty. However, the clinical utility of deleterious variants with moderate, uncertain penetrance depends more on assumed disutilities. The decision to include a gene on a panel depends on variant frequency, penetrance, and subjective utilities and should account for uncertainties around these factors.
Collapse
Affiliation(s)
- Jane W Liang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kurt D Christensen
- Center for Healthcare Research in Pediatrics, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
- Department of Population Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert C Green
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Mass General Brigham, Boston, MA, USA
- Ariadne Labs, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
3
|
Azuelos C, Marquis MA, Laberge AM. A systematic review of the assessment of the clinical utility of genomic sequencing: Implications of the lack of standard definitions and measures of clinical utility. Eur J Med Genet 2024; 68:104925. [PMID: 38432472 DOI: 10.1016/j.ejmg.2024.104925] [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: 09/21/2023] [Revised: 12/31/2023] [Accepted: 02/11/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE Exome sequencing (ES) and genome sequencing (GS) are diagnostic tests for rare genetic diseases. Studies report clinical utility of ES/GS. The goal of this systematic review is to establish how clinical utility is defined and measured in studies evaluating the impacts of ES/GS results for pediatric patients. METHODS Relevant articles were identified in PubMed, Medline, Embase, and Web of Science. Eligible studies assessed clinical utility of ES/GS for pediatric patients published before 2021. Other relevant articles were added based on articles' references. Articles were coded to assess definitions and measures of clinical utility. RESULTS Of 1346 articles, 83 articles met eligibility criteria. Clinical utility was not clearly defined in 19% of studies and 92% did not use an explicit measure of clinical utility. When present, definitions of clinical utility diverged from recommended definitions and varied greatly, from narrow (diagnostic yield of ES/GS) to broad (including decisions about withdrawal of care/palliative care and/or impacts on other family members). CONCLUSION Clinical utility is used to guide policy and practice decisions about test use. The lack of a standard definition of clinical utility of ES/GS may lead to under- or overestimations of clinical utility, complicating policymaking and raising ethical issues.
Collapse
Affiliation(s)
- Claudia Azuelos
- Medical Genetics, Dept of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada.
| | - Marc-Antoine Marquis
- Palliative Care, Dept of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada
| | - Anne-Marie Laberge
- Medical Genetics, Dept of Pediatrics, CHU Sainte-Justine and Université de Montréal, Canada.
| |
Collapse
|
4
|
Mighton C, Shickh S, Aguda V, Krishnapillai S, Adi-Wauran E, Bombard Y. From the patient to the population: Use of genomics for population screening. Front Genet 2022; 13:893832. [PMID: 36353115 PMCID: PMC9637971 DOI: 10.3389/fgene.2022.893832] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/26/2022] [Indexed: 10/22/2023] Open
Abstract
Genomic medicine is expanding from a focus on diagnosis at the patient level to prevention at the population level given the ongoing under-ascertainment of high-risk and actionable genetic conditions using current strategies, particularly hereditary breast and ovarian cancer (HBOC), Lynch Syndrome (LS) and familial hypercholesterolemia (FH). The availability of large-scale next-generation sequencing strategies and preventive options for these conditions makes it increasingly feasible to screen pre-symptomatic individuals through public health-based approaches, rather than restricting testing to high-risk groups. This raises anew, and with urgency, questions about the limits of screening as well as the moral authority and capacity to screen for genetic conditions at a population level. We aimed to answer some of these critical questions by using the WHO Wilson and Jungner criteria to guide a synthesis of current evidence on population genomic screening for HBOC, LS, and FH.
Collapse
Affiliation(s)
- Chloe Mighton
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Salma Shickh
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Vernie Aguda
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Centre for Medical Education, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Suvetha Krishnapillai
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Ella Adi-Wauran
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Yvonne Bombard
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Hayeems RZ, Bernier F, Boycott KM, Hartley T, Michaels-Igbokwe C, Marshall DA. Positioning whole exome sequencing in the diagnostic pathway for rare disease to optimise utility: a protocol for an observational cohort study and an economic evaluation. BMJ Open 2022; 12:e061468. [PMID: 36216418 PMCID: PMC9557316 DOI: 10.1136/bmjopen-2022-061468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Despite the superior diagnostic performance of exome and genome sequencing compared with conventional genetic tests, evidence gaps related to clinical utility and cost effectiveness have limited their availability in routine clinical practice in many jurisdictions. To inform adoption and reimbursement policy, this protocol provides a chain of evidence approach to determining the diagnostic utility, clinical utility and cost-effectiveness of whole exome sequencing (WES) from seven medical genetic centres in two Canadian provinces. METHODS AND ANALYSIS Using a multicentre observational cohort design, we will extract data specific to the pre-WES diagnostic pathway and 1-year post-WES medical management from electronic medical records for 650 patients with rare disease of suspected genetic aetiology who receive WES. The date from the clinical record will be linked to provincial administrative health database to capture healthcare resource use and estimate costs. Our analysis will: (1) define and describe diagnostic testing pathways that occur prior to WES among patients with rare disease, (2) determine the diagnostic utility of WES, characterised as the proportion of patients for whom causative DNA variants are identified, (3) determine the clinical utility of WES, characterised as a change in medical management triggered by WES results, (4) determine the pattern and cost of health service utilisation prior and 1 year following WES among patients who receive a diagnosis, do not receive a diagnosis, or receive an uncertain diagnosis and (5) estimate the cost-effectiveness of WES compared with conventional diagnostic testing pathways, measured by the incremental cost per additional patient diagnosed by WES using simulation modelling. ETHICS AND DISSEMINATION This protocol was approved by Clinical Trials Ontario (CTO-1577) and research ethics boards at the University of Calgary (REB18-0744 and REB20-1449) and University of Alberta (Pro0009156). Findings will be disseminated through academic publications and policy reports.
Collapse
Affiliation(s)
- Robin Z Hayeems
- Child Health Evaluative Sciences, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Francois Bernier
- Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kym M Boycott
- Department of Genetics, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Paediatrics, Facuty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Taila Hartley
- Department of Genetics, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Christine Michaels-Igbokwe
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Deborah A Marshall
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
6
|
Callahan KP, Mueller R, Flibotte J, Largent EA, Feudtner C. Measures of Utility Among Studies of Genomic Medicine for Critically Ill Infants: A Systematic Review. JAMA Netw Open 2022; 5:e2225980. [PMID: 35947384 PMCID: PMC9366540 DOI: 10.1001/jamanetworkopen.2022.25980] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
IMPORTANCE Genomic medicine holds promise to revolutionize care for critically ill infants by tailoring treatments for patients and providing additional prognostic information to families. However, measuring the utility of genomic medicine is not straightforward and has important clinical and ethical implications. OBJECTIVE To review the ways that researchers measure or neglect to measure the utility of genomic medicine for critically ill infants. EVIDENCE REVIEW This systematic review included prospective full-text studies of genomic medicine of both whole exome and genome sequencing in critically ill infants younger than 1 year. PubMed, Embase, Scopus, and Cochrane Library databases, the Cochrane Database of Systematic Reviews, and the ClinicalTrials.gov register were searched with an English language restriction for articles published from the inception of each database through May 2022. Search terms included variations of the following: gene, sequencing, intensive care, critical care, and infant. From the included articles, information on how utility was defined and measured was extracted and synthesized. Information was also extracted from patient cases that authors highlighted by providing additional information. Spearman rank-order correlation was used to evaluate the association between study size and utility. FINDINGS Synthesized data from the 21 included studies reflected results from 1654 patients. A mean of 46% (range, 15%-72%) of patients had a positive genetic test result, and a mean of 37% (range, 13%-61%) met the criteria for experiencing utility. Despite heterogeneity in how studies measured and reported utility, a standardized framework was created with 5 categories of utility: treatment change, redirection of care, prognostic information, reproductive information, and screening or subspecialty referral. Most studies omitted important categories of utility, notably personal utility (patient-reported benefits) (20 studies [95%]), utility of negative or uncertain results (15 [71%]), and disutility (harms) (20 [95%]). Studies disproportionally highlighted patient cases that resulted in treatment change. Larger studies reported substantially lower utility (r = -0.65; P = .002). CONCLUSIONS AND RELEVANCE This systematic review found that genomic medicine offered various categories of utility for a substantial proportion of critically ill infants. Studies measured utility in heterogeneous ways and focused more on documenting change than assessing meaningful benefit. Authors' decisions about which cases to highlight suggest that some categories of utility may be more important than others. A more complete definition of utility that is used consistently may improve understanding of potential benefits and harms of genetic medicine.
Collapse
Affiliation(s)
- Katharine Press Callahan
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Medical Ethics and Health Policy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Rebecca Mueller
- Department of Medical Ethics and Health Policy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - John Flibotte
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Emily A. Largent
- Department of Medical Ethics and Health Policy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Chris Feudtner
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Medical Ethics and Health Policy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
7
|
Krauss E, Macher J, Capasso J, Bernhardt B, Ali-KhanCatts Z, Levin A, Brandt R. Experiences of genetic testing among individuals with retinitis pigmentosa. Ophthalmic Genet 2022; 43:633-640. [PMID: 35796432 DOI: 10.1080/13816810.2022.2096243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Retinitis pigmentosa (RP) is a genetically heterogeneous retinal dystrophy which results in progressive vision loss. There is scant literature on the experiences of genetic testing in patients with RP. MATERIALS AND METHODS Patients with a clinical diagnosis of RP who received genetic testing at the Wills Eye Ocular Genetics clinic between 2016 and 2020 were recruited. Telephone interviews were conducted using a semi-structured guide designed to elicit participant experiences with genetic testing. A thematic analysis was performed to describe patterns in participant responses. RESULTS Twelve patients participated. Seven participants identified as female and five as male, with ages ranging from 22 to 70. Ten patients had positive genetic test results, while two had negative genetic testing. Reported motivations for genetic testing included qualification for clinical trials (58% of total participants), determination of etiology or usal gene (50%), reproductive concerns (50%), and prognostic outlook (50%). Most participants (75%) expressed satisfaction about their decision to pursue genetic testing. Participants with both positive and negative genetic testing reported persistent uncertainty regarding their prognosis for visual decline (50%). Genetic confirmation of disease leads to initiation of safety and vision-protecting health behaviors (42%). CONCLUSION Patients with RP are generally satisfied with their testing experience, despite approaching testing with a wide range of motivations and expectations. Future research can leverage this methodology to identify targets for improvement in pre- and post-test education and counselling.
Collapse
Affiliation(s)
- Emily Krauss
- Duke Eye Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jared Macher
- University of Rochester School of Medicine, Rochester, New York, USA
| | - Jenina Capasso
- Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, Rochester, New York, USA.,Pediatric Genetics, Golisano Children's Hospital, Rochester, New York, USA
| | - Barbara Bernhardt
- Human Genetics and Genetic Counseling, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Zohra Ali-KhanCatts
- Human Genetics and Genetic Counseling, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alex Levin
- Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, Rochester, New York, USA.,Pediatric Genetics, Golisano Children's Hospital, Rochester, New York, USA
| | - Rachael Brandt
- Human Genetics and Genetic Counseling, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Lankenau Medical Center, Main Line Health Hospitals, Wynnewood, Pennsylvania, USA.,Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, USA
| |
Collapse
|
8
|
Shickh S, Mighton C, Clausen M, Adi-Wauran E, Hirjikaka D, Kodida R, Krishnapillai S, Reble E, Sam J, Shaw A, Lerner-Ellis J, Baxter NN, Laupacis A, Bombard Y. "Doctors shouldn't have to cheat the system": Clinicians' real-world experiences of the utility of genomic sequencing. Genet Med 2022; 24:1888-1898. [PMID: 35612591 DOI: 10.1016/j.gim.2022.04.024] [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: 02/23/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Emerging genetic tests such as genomic sequencing (GS) can generate a broad range of benefits, but funding criteria only prioritize diagnosis and clinical management. There is limited evidence on all types of benefits obtained from GS in practice. We aimed to explore real-world experiences of Canadian clinicians across specialties on the full range of benefits obtained from the results from GS. METHODS We conducted a qualitative study using semistructured interviews with Canadian clinicians. Transcripts were thematically analyzed using constant comparison. RESULTS In total, 25 clinicians participated, including 12 geneticists, 7 genetic counselors, 4 oncologists, 1 neurologist, and 1 family physician. Although diagnoses and management were the most valued benefits of GS, clinicians also prioritized nontraditional utility, such as access to community supports. However, clinicians felt "restricted" by funding bodies, which only approved funding when GS would inform diagnoses and management. Consequently, clinicians sought ways to "cheat the system" to access GS (eg, research testing) but acknowledged workarounds were burdensome, drove inequity, and undermined patient care. CONCLUSION Current governance structures undervalue real-world benefits of GS leading clinicians to adopt workarounds, which jeopardize patient care. These results support calls for the expansion of the definition of clinical utility and research to quantify the additional benefits.
Collapse
Affiliation(s)
- Salma Shickh
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Chloe Mighton
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Marc Clausen
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Ella Adi-Wauran
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Daena Hirjikaka
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Rita Kodida
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Suvetha Krishnapillai
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Emma Reble
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Jordan Sam
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Angela Shaw
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Jordan Lerner-Ellis
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nancy N Baxter
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Andreas Laupacis
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Yvonne Bombard
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
The Clinician-reported Genetic testing Utility InDEx (C-GUIDE): Preliminary evidence of validity and reliability. Genet Med 2021; 24:430-438. [PMID: 34906486 DOI: 10.1016/j.gim.2021.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Demonstrating the clinical utility of genetic testing is fundamental to clinical adoption and reimbursement, but standardized definitions and measurement strategies for this construct do not exist. The Clinician-reported Genetic testing Utility InDEx (C-GUIDE) offers a novel measure to fill this gap. This study assessed its validity and inter-rater reliability. METHODS Genetics professionals completed C-GUIDE after disclosure of test results to patients. Construct validity was assessed using regression analysis to measure associations between C-GUIDE and global item scores as well as potentially explanatory variables. Inter-rater reliability was assessed by administering a vignette-based survey to genetics professionals and calculating Krippendorff's α. RESULTS On average, a 1-point increase in the global item score was associated with an increase of 3.0 in the C-GUIDE score (P < .001). Compared with diagnostic results, partially/potentially diagnostic and nondiagnostic results were associated with a reduction in C-GUIDE score of 9.5 (P < .001) and 10.2 (P < .001), respectively. Across 19 vignettes, Krippendorff's α was 0.68 (95% CI: 0.63-0.72). CONCLUSION C-GUIDE showed acceptable validity and inter-rater reliability. Although further evaluation is required, C-GUIDE version 1.2 can be useful as a standardized approach to assess the clinical utility of genetic testing.
Collapse
|
10
|
Shickh S, Mighton C, Uleryk E, Pechlivanoglou P, Bombard Y. The clinical utility of exome and genome sequencing across clinical indications: a systematic review. Hum Genet 2021; 140:1403-1416. [PMID: 34368901 DOI: 10.1007/s00439-021-02331-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022]
Abstract
Exome sequencing and genome sequencing have the potential to improve clinical utility for patients undergoing genetic investigations. However, evidence of clinical utility is limited to pediatric populations; we aimed to fill this gap by conducting a systematic review of the literature on the clinical utility of exome/genome sequencing across disease indications in pediatric and adult populations. MEDLINE, EMBASE and Cochrane Library were searched between 2016 and 2020. Quantitative studies evaluating diagnostic yield were included; other measures of clinical utility such as changes to clinical management were documented if reported. Two reviewers screened, extracted data, and appraised risk of bias. Fifty studies met our inclusion criteria. All studies reported diagnostic yield, which ranged from 3 to 70%, with higher range of yields reported for neurological indications and acute illness ranging from 22 to 68% and 37-70%, respectively. Diagnoses triggered a range of clinical management changes including surveillance, reproductive-risk counseling, and identifying at-risk relatives in 4-100% of patients, with higher frequencies reported for acute illness ranging from 67 to 95%. The frequency of variants of uncertain significance ranged from 5 to 85% across studies with a potential trend of decreasing frequency over time and higher rates identified in patients of non-European ancestry. This review provides evidence for a higher range of diagnostic yield of exome/genome sequencing compared to standard genetic tests, particularly in neurological and acute indications. However, we identified significant heterogeneity in study procedures and outcomes, precluding a meaningful meta-analysis and certainty in the evidence available for decision-making. Future research that incorporates a comprehensive and consistent approach in capturing clinical utility of exome/genome sequencing across broader ancestral groups is necessary to improve diagnostic accuracy and yield and allow for analysis of trends over time.Prospero registration CRD42019094101.
Collapse
Affiliation(s)
- Salma Shickh
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.,Genomics Health Services Research Program, Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, ON, Canada
| | - Chloe Mighton
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.,Genomics Health Services Research Program, Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, ON, Canada
| | | | - Petros Pechlivanoglou
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.,The Hospital for Sick Children, Toronto, ON, Canada
| | - Yvonne Bombard
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada. .,Genomics Health Services Research Program, Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, ON, Canada. .,Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, 30 Bond Street, Toronto, ON, M5B 1W8, Canada.
| |
Collapse
|
11
|
Walcott SE, Miller FA, Dunsmore K, Lazor T, Feldman BM, Hayeems RZ. Measuring clinical utility in the context of genetic testing: a scoping review. Eur J Hum Genet 2021; 29:378-386. [PMID: 33087880 PMCID: PMC7940410 DOI: 10.1038/s41431-020-00744-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 01/20/2023] Open
Abstract
Standardized approaches to measuring clinical utility will enable more robust evaluations of genetic tests. To characterize how clinical utility has been measured, this scoping review examined outcomes used to operationalize this concept in the context of genetic testing, spanning relevant literature (2015-2017). The search strategy and analysis were guided by the Fryback and Thornbury hierarchical model of efficacy (FT Model). Through searches in Ovid MEDLINE, EMBASE and Web of Science, 194 publications were identified for inclusion. Two coders reviewed titles, abstracts, and full texts to determine eligibility. Results were analyzed using thematic and frequency analyses. This review generated a catalog of outcomes mapped to the efficacy domains of the FT Model. The degree of representation observed in each domain varied by the clinical purpose and clinical indication of genetic testing. Diagnostic accuracy (68%), technical (28.4%), and patient outcome (28.4%) efficacy studies were represented at the highest rate. Findings suggest that the FT Model is suitable for the genetics context however domain refinements may be warranted. More diverse clinical settings, robust study designs, and novel strategies for measuring clinical utility are needed.
Collapse
Affiliation(s)
- Shantel E Walcott
- Institute of Health Policy Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Fiona A Miller
- Institute of Health Policy Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Toronto Health Economics and Technology Assessment, Toronto, ON, Canada
| | - Kourtney Dunsmore
- Institute of Health Policy Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tanya Lazor
- Institute of Health Policy Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Brian M Feldman
- Institute of Health Policy Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Program in Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Paediatrics, Division of Rheumatology, University of Toronto, Toronto, ON, Canada
| | - Robin Z Hayeems
- Institute of Health Policy Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
- Program in Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, ON, Canada.
| |
Collapse
|
12
|
Primiero CA, Yanes T, Finnane A, Soyer HP, McInerney-Leo AM. A Systematic Review on the Impact of Genetic Testing for Familial Melanoma I: Primary and Secondary Preventative Behaviours. Dermatology 2021; 237:806-815. [PMID: 33588421 DOI: 10.1159/000513919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/12/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Increasing availability of panel testing for known high-penetrance familial melanoma genes has made it possible to improve risk awareness in those at greatest risk. Prior to wider implementation, the role of genetic testing in preventing melanoma, through influencing primary and secondary preventative behaviours, requires clarification. METHODS Database searches of PubMed, Embase, CINAHL, PsycINFO and the Cochrane Library were conducted for studies describing preventative behaviour outcomes in response to genetic testing for melanoma risk. Publications describing original research of any study type were screened for eligibility. RESULTS Eighteen publications describing 11 unique studies were reviewed. Outcomes assessed are based on health behaviour recommendations for those at increased risk: adherence to sun-protective behaviour (SPB); clinical skin examinations (CSE); skin self-examinations (SSE); and family discussion of risk. Overall, modest increases in adherence to primary prevention strategies of SPB were observed following genetic testing. Importantly, there were no net decreases in SPB found amongst non-carriers. For secondary preventative behaviour outcomes, including CSE and SSE, increases in post-test intentions and long-term adherence were reported across several subgroups in approximately half of the studies. While this increase reached significance in mutation carriers in some studies, one study reported a significant decline in annual CSE adherence of non-mutation carriers. CONCLUSIONS Evidence reviewed suggests that genetic testing has a modestly positive impact on preventative behaviour in high-risk individuals. Furthermore, improvements are observed regardless of mutation carrier status, although greater adherence is found in carriers. While additional studies of more diverse cohorts would be needed to inform clinical recommendations, the findings are encouraging and suggest that genetic testing for melanoma has a positive impact on preventative behaviours.
Collapse
Affiliation(s)
- Clare A Primiero
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Queensland, Australia
| | - Tatiane Yanes
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Queensland, Australia
| | - Anna Finnane
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| | - H Peter Soyer
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Queensland, Australia,
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia,
| | - Aideen M McInerney-Leo
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, Queensland, Australia
| |
Collapse
|
13
|
Malsagova KA, Butkova TV, Kopylov AT, Izotov AA, Potoldykova NV, Enikeev DV, Grigoryan V, Tarasov A, Stepanov AA, Kaysheva AL. Pharmacogenetic Testing: A Tool for Personalized Drug Therapy Optimization. Pharmaceutics 2020; 12:E1240. [PMID: 33352764 PMCID: PMC7765968 DOI: 10.3390/pharmaceutics12121240] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
Pharmacogenomics is a study of how the genome background is associated with drug resistance and how therapy strategy can be modified for a certain person to achieve benefit. The pharmacogenomics (PGx) testing becomes of great opportunity for physicians to make the proper decision regarding each non-trivial patient that does not respond to therapy. Although pharmacogenomics has become of growing interest to the healthcare market during the past five to ten years the exact mechanisms linking the genetic polymorphisms and observable responses to drug therapy are not always clear. Therefore, the success of PGx testing depends on the physician's ability to understand the obtained results in a standardized way for each particular patient. The review aims to lead the reader through the general conception of PGx and related issues of PGx testing efficiency, personal data security, and health safety at a current clinical level.
Collapse
Affiliation(s)
- Kristina A. Malsagova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 109028 Moscow, Russia; (T.V.B.); (A.T.K.); (A.A.I.); (A.A.S.); (A.L.K.)
| | - Tatyana V. Butkova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 109028 Moscow, Russia; (T.V.B.); (A.T.K.); (A.A.I.); (A.A.S.); (A.L.K.)
| | - Arthur T. Kopylov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 109028 Moscow, Russia; (T.V.B.); (A.T.K.); (A.A.I.); (A.A.S.); (A.L.K.)
| | - Alexander A. Izotov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 109028 Moscow, Russia; (T.V.B.); (A.T.K.); (A.A.I.); (A.A.S.); (A.L.K.)
| | - Natalia V. Potoldykova
- Institute of Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (N.V.P.); (D.V.E.); (V.G.)
| | - Dmitry V. Enikeev
- Institute of Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (N.V.P.); (D.V.E.); (V.G.)
| | - Vagarshak Grigoryan
- Institute of Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (N.V.P.); (D.V.E.); (V.G.)
| | - Alexander Tarasov
- Institute of Linguistics and Intercultural Communication, Sechenov University, 119992 Moscow, Russia;
| | - Alexander A. Stepanov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 109028 Moscow, Russia; (T.V.B.); (A.T.K.); (A.A.I.); (A.A.S.); (A.L.K.)
| | - Anna L. Kaysheva
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 109028 Moscow, Russia; (T.V.B.); (A.T.K.); (A.A.I.); (A.A.S.); (A.L.K.)
| |
Collapse
|
14
|
Hayeems RZ, Dimmock D, Bick D, Belmont JW, Green RC, Lanpher B, Jobanputra V, Mendoza R, Kulkarni S, Grove ME, Taylor SL, Ashley E. Clinical utility of genomic sequencing: a measurement toolkit. NPJ Genom Med 2020; 5:56. [PMID: 33319814 PMCID: PMC7738524 DOI: 10.1038/s41525-020-00164-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022] Open
Abstract
Whole-genome sequencing (WGS) is positioned to become one of the most robust strategies for achieving timely diagnosis of rare genomic diseases. Despite its favorable diagnostic performance compared to conventional testing strategies, routine use and reimbursement of WGS are hampered by inconsistencies in the definition and measurement of clinical utility. For example, what constitutes clinical utility for WGS varies by stakeholder's perspective (physicians, patients, families, insurance companies, health-care organizations, and society), clinical context (prenatal, pediatric, critical care, adult medicine), and test purpose (diagnosis, screening, treatment selection). A rapidly evolving technology landscape and challenges associated with robust comparative study design in the context of rare disease further impede progress in this area of empiric research. To address this challenge, an expert working group of the Medical Genome Initiative was formed. Following a consensus-based process, we align with a broad definition of clinical utility and propose a conceptually-grounded and empirically-guided measurement toolkit focused on four domains of utility: diagnostic thinking efficacy, therapeutic efficacy, patient outcome efficacy, and societal efficacy. For each domain of utility, we offer specific indicators and measurement strategies. While we focus on diagnostic applications of WGS for rare germline diseases, this toolkit offers a flexible framework for best practices around measuring clinical utility for a range of WGS applications. While we expect this toolkit to evolve over time, it provides a resource for laboratories, clinicians, and researchers looking to characterize the value of WGS beyond the laboratory.
Collapse
Affiliation(s)
- Robin Z Hayeems
- Program in Child Health Evaluative Sciences, The Hospital for Sick Children and the Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, ON, Canada.
| | - David Dimmock
- Rady Children's Hospital Institute for Genomic Medicine, San Diego, CA, USA
| | - David Bick
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - Robert C Green
- Brigham and Women's Hospital Broad Institute and Harvard Medical School, Boston, MA, USA
| | | | - Vaidehi Jobanputra
- New York Genome Center, New York, NY, USA
- Department of Pathology and Cell Biology Columbia University Medical Center, New York, NY, USA
| | - Roberto Mendoza
- The Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shashi Kulkarni
- Baylor Genetics and Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | | | | |
Collapse
|
15
|
Elson SL, Furlotte NA, Hromatka BS, Wilson CH, Mountain JL, Rowbotham HM, Varga EA, Francke U. Direct-to-consumer genetic testing for factor V Leiden and prothrombin 20210G>A: the consumer experience. Mol Genet Genomic Med 2020; 8:e1468. [PMID: 32940023 PMCID: PMC7667316 DOI: 10.1002/mgg3.1468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 01/19/2023] Open
Abstract
Background Clinical genetic testing for inherited predisposition to venous thromboembolism (VTE) is common among patients and their families. However, there is incomplete consensus about which individuals should receive testing, and the relative risks and benefits. Methods We assessed outcomes of receiving direct‐to‐consumer (DTC) results for the two most common genetic risk factors for VTE, factor V Leiden in the F5 gene (FVL) and prothrombin 20210G>A in the F2 gene (PT). Two thousand three hundred fifty‐four customers (1244 variant‐positive and 1110 variant‐negative individuals) of the personal genetics company 23andMe, Inc., who had received results online for F5 and F2 variants, participated in an online survey‐based study. Participants responded to questions about perception of VTE risk, discussion of results with healthcare providers (HCPs) and recommendations received, actions taken to control risk, emotional responses to receiving risk results, and perceived value of the information. Results Most participants (90% of variant‐positive individuals, 99% of variant‐negative individuals) had not previously been tested for F5 and/or F2 variants. The majority of variant‐positive individuals correctly perceived that they were at higher than average risk for developing VTE. These individuals reported moderate rates of discussing results with HCPs (41%); receiving prevention advice from HCPs (31%), and making behavioral changes to control risk (e.g., exercising more, 30%). A minority (36%) of variant‐positive individuals worried more after receiving VTE results. Nevertheless, most participants reported that knowing their risk had been an advantage (78% variant‐positive and 58% variant‐negative) and were satisfied knowing their genetic probability for VTE (81% variant‐positive and 67% variant‐negative). Conclusion Consumers reported moderate rates of behavioral change and perceived personal benefit from receiving DTC genetic results for VTE risk. We assessed the consumer experience receiving direct‐to‐consumer (DTC) genetic risk for venous thromboembolism (VTE). 2354 customers (1244 variant‐positive and 1110 variant‐negative individuals) of the personal genetics company 23andMe, Inc., who had received results online for risk variants in clotting factor genes F2 and F5, participated in an online survey‐based study. Participants responded to questions about perception of VTE risk, discussion of results with healthcare providers and recommendations received, actions taken to control risk, emotional responses to receiving risk results, and perceived value of the information.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Uta Francke
- 23andMe, Inc, Sunnyvale, CA, USA.,Department of Genetics, Stanford University Stanford, CA, USA
| |
Collapse
|
16
|
Bíró K, Dombrádi V, Fekete Z, Bányai G, Boruzs K, Nagy A, Ádány R. Investigating the knowledge of and public attitudes towards genetic testing within the Visegrad countries: a cross-sectional study. BMC Public Health 2020; 20:1380. [PMID: 32912246 PMCID: PMC7488256 DOI: 10.1186/s12889-020-09473-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/30/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Previous studies have investigated various factors that can determine the attitudes of the citizens considering genetic testing. However, none of them investigated how these attitudes may differ between the Visegrad countries. METHODS In this cross-sectional study a questionnaire developed by Dutch researchers was translated and used in Hungary, Slovakia, Czechia and Poland. In each country 1000 adult citizens were asked on the topics of personal benefits regarding genetic tests, genetic determinism, and finally, the availability and usage of genetic testing. Multivariate robust regression model was created including several possible influencing factors (such as age, sex, education, marital status, religiousness, and having a genetic test within the nuclear family) to identify the possible differences between the four countries. RESULTS The Hungarian citizens had the most positive opinion on the personal benefits of genetic testing followed by the Czech, Slovak and Polish. All differences were significant in this regard. Considering genetic determinism, the Slovak citizens had a significantly firmer belief in this issue compared to the Hungarians. No other significant differences were observed in this domain. On the topic of the availability and use of genetic testing the Hungarian citizens had the most accepting opinion among the four countries, followed by the Czech citizens. In this domain the Polish and Slovak answers did not differ significantly from each other. CONCLUSIONS Significant differences were observed even when considering various confounding effects. As the underlying reasons for these discrepancies are unknown, future studies should investigate this enigma among the four countries.
Collapse
Affiliation(s)
- Klára Bíró
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Viktor Dombrádi
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Zita Fekete
- Department of Behavioural Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Bányai
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Klára Boruzs
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Attila Nagy
- Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Róza Ádány
- MTA-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
17
|
Mighton C, Carlsson L, Clausen M, Casalino S, Shickh S, McCuaig L, Joshi E, Panchal S, Semotiuk K, Ott K, Elser C, Eisen A, Kim RH, Lerner-Ellis J, Carroll JC, Glogowski E, Schrader K, Bombard Y. Quality of life drives patients' preferences for secondary findings from genomic sequencing. Eur J Hum Genet 2020; 28:1178-1186. [PMID: 32424322 PMCID: PMC7609335 DOI: 10.1038/s41431-020-0640-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/31/2020] [Accepted: 04/14/2020] [Indexed: 01/01/2023] Open
Abstract
There is growing impetus to include measures of personal utility, the nonmedical value of information, in addition to clinical utility in health technology assessment (HTA) of genomic tests such as genomic sequencing (GS). However, personal utility and clinical utility are challenging to define and measure. This study aimed to explore what drives patients' preferences for hypothetically learning medically actionable and non-medically actionable secondary findings (SF), capturing clinical and personal utility; this may inform development of measures to evaluate patient outcomes following return of SF. Semi-structured interviews were conducted with adults with a personal or family cancer history participating in a trial of a decision aid for selection of SF from genomic sequencing (GS) ( www.GenomicsADvISER.com ). Interviews were analyzed thematically using constant comparison. Preserving health-related and non-health-related quality of life was an overarching motivator for both learning and not learning SF. Some participants perceived that learning SF would help them "have a good quality of life" through informing actions to maintain physical health or leading to psychological benefits such as emotional preparation for disease. Other participants preferred not to learn SF because results "could ruin your quality of life," such as by causing negative psychological impacts. Measuring health-related and non-health-related quality of life may capture outcomes related to clinical and personal utility of GS and SF, which have previously been challenging to measure. Without appropriate measures, generating and synthesizing evidence to evaluate genomic technologies such as GS will continue to be a challenge, and will undervalue potential benefits of GS and SF.
Collapse
Affiliation(s)
- Chloe Mighton
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada
| | - Lindsay Carlsson
- Lawrence S. Bloomberg Faculty of Nursing, University of Toronto, Toronto, ON, Canada
- University Health Network, Toronto, ON, Canada
| | - Marc Clausen
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada
| | - Selina Casalino
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada
| | - Salma Shickh
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada
| | - Laura McCuaig
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Esha Joshi
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada
| | | | | | - Karen Ott
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Christine Elser
- University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Sinai Health System, Toronto, ON, Canada
| | - Andrea Eisen
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Raymond H Kim
- University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Sinai Health System, Toronto, ON, Canada
| | - Jordan Lerner-Ellis
- Sinai Health System, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - June C Carroll
- Sinai Health System, Toronto, ON, Canada
- Department of Family & Community Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Kasmintan Schrader
- BC Cancer Agency, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Yvonne Bombard
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health, Toronto, ON, Canada.
| |
Collapse
|
18
|
Harmonizing gene signatures to predict benefit from adjuvant chemotherapy in early breast cancer. Curr Opin Oncol 2020; 31:472-479. [PMID: 31593974 DOI: 10.1097/cco.0000000000000570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Breast cancer is a heterogeneous disease, including different subtypes with their own biology, prognosis, clinical characteristics and treatment. To date, traditional clinical and pathological determinants remain the main factors guiding treatment decision-making; however, the development of multigene assays improved the ability to predict the risk of recurrence in patients with early-stage breast cancer. These tools underwent an extensive independent validation and have already been partly incorporated into clinical practice. RECENT FINDINGS The current article summarizes current evidence for the use of the different genomic assays in clinical practice, their characteristics and validation studies. A few studies comparing available genomic assays revealed that they provide different information with a modest correlation and that they are not interchangeable; other trials are currently ongoing in this setting. SUMMARY Variability across different gene signatures may be a challenge for the optimal management of the individual patient, hence each assay should be used for the clinical setting in which it has been validated.
Collapse
|
19
|
Zebrowski AM, Ellis DE, Barg FK, Sperber NR, Bernhardt BA, Denny JC, Dexter PR, Ginsburg GS, Horowitz CR, Johnson JA, Levy MA, Orlando LA, Pollin TI, Skaar TC, Kimmel SE. Qualitative study of system-level factors related to genomic implementation. Genet Med 2019; 21:1534-1540. [PMID: 30467402 PMCID: PMC6533158 DOI: 10.1038/s41436-018-0378-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/09/2018] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Research on genomic medicine integration has focused on applications at the individual level, with less attention paid to implementation within clinical settings. Therefore, we conducted a qualitative study using the Consolidated Framework for Implementation Research (CFIR) to identify system-level factors that played a role in implementation of genomic medicine within Implementing GeNomics In PracTicE (IGNITE) Network projects. METHODS Up to four study personnel, including principal investigators and study coordinators from each of six IGNITE projects, were interviewed using a semistructured interview guide that asked interviewees to describe study site(s), progress at each site, and factors facilitating or impeding project implementation. Interviews were coded following CFIR inner-setting constructs. RESULTS Key barriers included (1) limitations in integrating genomic data and clinical decision support tools into electronic health records, (2) physician reluctance toward genomic research participation and clinical implementation due to a limited evidence base, (3) inadequate reimbursement for genomic medicine, (4) communication among and between investigators and clinicians, and (5) lack of clinical and leadership engagement. CONCLUSION Implementation of genomic medicine is hindered by several system-level barriers to both research and practice. Addressing these barriers may serve as important facilitators for studying and implementing genomics in practice.
Collapse
Affiliation(s)
- Alexis M Zebrowski
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, USA
| | - Darcy E Ellis
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Frances K Barg
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Family Medicine and Community Health, University of Pennsylvania, Philadelphia, PA, USA
| | - Nina R Sperber
- Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Barbara A Bernhardt
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of General Internal Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Paul R Dexter
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Geoffrey S Ginsburg
- Duke Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC, USA
| | - Carol R Horowitz
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Mia A Levy
- Departments of Biomedical Informatics and Medicine, Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lori A Orlando
- Duke Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC, USA
- Division of General Internal Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Toni I Pollin
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Todd C Skaar
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen E Kimmel
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA.
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Medicine, Pearlman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Briggs S, Slade I. Evaluating the Integration of Genomics into Cancer Screening Programmes: Challenges and Opportunities. CURRENT GENETIC MEDICINE REPORTS 2019; 7:63-74. [PMID: 32117599 PMCID: PMC7019642 DOI: 10.1007/s40142-019-00162-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW As the costs of genomic testing have fallen, and our understanding of genetic susceptibility to cancers has grown, there has been increasing interest in incorporating testing for cancer susceptibility genes, and polygenic risk estimates, into population cancer screening. A growing body of evidence suggests that this would be both clinically and cost-effective. In this article, we aim to explore the frameworks used to evaluate screening programmes, evaluate whether population screening for cancer susceptibility can be assessed using these standards, and consider additional issues and outcomes of importance in this context. RECENT FINDINGS There are tensions between traditional approaches of genetic testing (utilising tests with high sensitivity and specificity) and the principles of population screening (in which the screening test typically has low specificity), as well as the frameworks used to evaluate the two. Despite the existence of many screening guidelines, including consensus papers, these often do not align fully with broader considerations of genetic test evaluation. Population screening for genetic risk in cancer shifts the focus from diagnostics to prognostication and has wider implications for personal and familial health than existing screening programmes. In addition, understanding of the prevalence and penetrance of cancer susceptibility genes, required by many screening guidelines, may only be obtainable through population-level testing; prospective multi-disciplinary research alongside implementation will be essential. SUMMARY Appropriate evaluation of genetic screening for cancer risk will require modification of existing screening frameworks to incorporate additional complexity of outcomes and population values. As evidence supporting population screening for cancer susceptibility mounts, development of an appropriate evaluative framework, and expansion of public dialogue will be key to informing policy.
Collapse
Affiliation(s)
- Sarah Briggs
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Ingrid Slade
- Wellcome Centre for Ethics and Humanities and Ethox Centre, Nuffield Department of Population Health, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Oxford, OX3 7LF UK
| |
Collapse
|
21
|
Smith HS, Swint JM, Lalani SR, Yamal JM, de Oliveira Otto MC, Castellanos S, Taylor A, Lee BH, Russell HV. Clinical Application of Genome and Exome Sequencing as a Diagnostic Tool for Pediatric Patients: a Scoping Review of the Literature. Genet Med 2019; 21:3-16. [PMID: 29760485 DOI: 10.1038/s41436-018-0024-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Availability of clinical genomic sequencing (CGS) has generated questions about the value of genome and exome sequencing as a diagnostic tool. Analysis of reported CGS application can inform uptake and direct further research. This scoping literature review aims to synthesize evidence on the clinical and economic impact of CGS. METHODS PubMed, Embase, and Cochrane were searched for peer-reviewed articles published between 2009 and 2017 on diagnostic CGS for infant and pediatric patients. Articles were classified according to sample size and whether economic evaluation was a primary research objective. Data on patient characteristics, clinical setting, and outcomes were extracted and narratively synthesized. RESULTS Of 171 included articles, 131 were case reports, 40 were aggregate analyses, and 4 had a primary economic evaluation aim. Diagnostic yield was the only consistently reported outcome. Median diagnostic yield in aggregate analyses was 33.2% but varied by broad clinical categories and test type. CONCLUSION Reported CGS use has rapidly increased and spans diverse clinical settings and patient phenotypes. Economic evaluations support the cost-saving potential of diagnostic CGS. Multidisciplinary implementation research, including more robust outcome measurement and economic evaluation, is needed to demonstrate clinical utility and cost-effectiveness of CGS.
Collapse
Affiliation(s)
- Hadley Stevens Smith
- Baylor College of Medicine, The University of Texas School of Public Health, Houston, Texas, USA
| | - J Michael Swint
- The University of Texas School of Public Health, The Center for Clinical Research and Evidence-Based Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Seema R Lalani
- Baylor College of Medicine, Baylor Genetics Laboratory, Houston, Texas, USA
| | - Jose-Miguel Yamal
- The University of Texas School of Public Health, Houston, Texas, USA
| | | | | | - Amy Taylor
- Texas Medical Center Library, Houston, Texas, USA
| | | | - Heidi V Russell
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
22
|
Pereira S, Robinson JO, Gutierrez AM, Petersen DK, Hsu RL, Lee CH, Schwartz TS, Holm IA, Beggs AH, Green RC, McGuire AL. Perceived Benefits, Risks, and Utility of Newborn Genomic Sequencing in the BabySeq Project. Pediatrics 2019; 143:S6-S13. [PMID: 30600265 PMCID: PMC6480393 DOI: 10.1542/peds.2018-1099c] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/03/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES There is interest in applying genomic sequencing (GS) to newborns' clinical care. Here we explore parents' and clinicians' attitudes toward and perceptions of the risks, benefits, and utility of newborn GS compared with newborn screening (NBS) prior to receiving study results. METHODS The BabySeq Project is a randomized controlled trial used to explore the impact of integrating GS into the clinical care of newborns. Parents (n = 493) of enrolled infants (n = 309) and clinicians (n = 144) completed a baseline survey at enrollment. We examined between-group differences in perceived utility and attitudes toward NBS and GS. Open-ended responses about risks and benefits of each technology were categorized by theme. RESULTS The majority of parents (71%) and clinicians (51%) agreed that there are health benefits of GS, although parents and clinicians agreed more that there are risks associated with GS (35%, 70%) than with NBS (19%, 39%; all P < .05). Parents perceived more benefit and less risk of GS than did clinicians. Clinicians endorsed concerns about privacy and discrimination related to genomic information more strongly than did parents, and parents anticipated benefits of GS that clinicians did not. CONCLUSIONS Parents and clinicians are less confident in GS than NBS, but parents perceive a more favorable risk/benefit ratio of GS than do clinicians. Clinicians should be aware that parents' optimism may stem from their perceived benefits beyond clinical utility.
Collapse
Affiliation(s)
- Stacey Pereira
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas
| | - Jill Oliver Robinson
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas
| | - Amanda M Gutierrez
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas
| | - Devan K Petersen
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas
| | - Rebecca L Hsu
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas
| | - Caroline H Lee
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas
| | - Talia S Schwartz
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital
- Department of Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Ingrid A Holm
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital
- Department of Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital
- Department of Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Robert C Green
- Department of Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts
- Partners Healthcare Personalized Medicine, Cambridge, Massachusetts; and
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Amy L McGuire
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, Texas;
| |
Collapse
|
23
|
Viberg Johansson J, Segerdahl P, Ugander UH, Hansson MG, Langenskiöld S. Making sense of genetic risk: A qualitative focus-group study of healthy participants in genomic research. PATIENT EDUCATION AND COUNSELING 2018; 101:422-427. [PMID: 28947362 DOI: 10.1016/j.pec.2017.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE It is well known that research participants want to receive genetic risk information that is about high risks, serious diseases and potential preventive measures. The aim of this study was to explore, by qualitative means, something less well known: how do healthy research participants themselves make sense of genetic risk information? METHOD A phenomenographic approach was chosen to explore research participants' understanding and assessment of genetic risk. We conducted four focus-group (N=16) interviews with participants in a research programme designed to identify biomarkers for cardiopulmonary disease. RESULTS Among the research participants, we found four ways of understanding genetic risk: as a binary concept, as an explanation, as revealing who I am (knowledge of oneself) and as affecting life ahead. CONCLUSION Research participants tend to understand genetic risk as a binary concept. This does not necessarily imply a misunderstanding of, or an irrational approach to, genetic risk. Rather, it may have a heuristic function in decision-making. PRACTICAL IMPLICATIONS Risk communication may be enhanced by tailoring the communication to the participants' own lay conceptions. For example, researchers and counselors should address risk in binary terms, maybe looking out for how individual participants search for threshold figures.
Collapse
Affiliation(s)
- Jennifer Viberg Johansson
- Centre for Research Ethics & Bioethics, Department of Public Health and Caring Sciences, Uppsala University, Box 564, Uppsala SE-751 22, Sweden.
| | - Pär Segerdahl
- Centre for Research Ethics & Bioethics, Department of Public Health and Caring Sciences, Uppsala University, Box 564, Uppsala SE-751 22, Sweden
| | | | - Mats G Hansson
- Centre for Research Ethics & Bioethics, Department of Public Health and Caring Sciences, Uppsala University, Box 564, Uppsala SE-751 22, Sweden
| | - Sophie Langenskiöld
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden; Department of Learning, Informatics, Management and Ethics, Medical Management Centre, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Abstract
Breast cancer is a heterogeneous disease, with different subtypes having a distinct biological, molecular, and clinical course. Assessments of standard clinical and pathological features have traditionally been used to determine the use of adjuvant systemic therapy in patients with early stage breast cancer; however, the ability to identify those who will benefit from adjuvant chemotherapy remains a challenge, leading to the overtreatment of some patients. Advances in molecular medicine have substantially improved the accuracy of gene-expression profiling of breast tumours, resulting in improvements in the ability to predict a patient's risk of breast cancer recurrence and likely response to endocrine therapy and/or chemotherapy. These genomic assays, several of which are commercially available, have aided physicians in tailoring treatment decisions for patients at the individual level. Herein, we describe the available data on the clinical validity of the most widely available assays in patients with early stage breast cancer, with a focus on the development, validation, and clinical application of these assays, in addition to the anticipated outcomes of ongoing prospective trials. We also review data from comparative studies of these assays and from cost-effectiveness analyses relating to their clinical use.
Collapse
|
25
|
Malek J, Slashinski MJ, Robinson JO, Gutierrez AM, Parsons DW, Plon SE, McCullough LB, McGuire AL. Parental Perspectives on Whole Exome Sequencing in Pediatric Cancer: A Typology of Perceived Utility. JCO Precis Oncol 2017; 1. [PMID: 31886451 DOI: 10.1200/po.17.00010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose To explore how parents of pediatric cancer patients perceived the utility of clinical tumor and germline whole-exome sequencing (WES) results. Patients and Methods We conducted longitudinal interviews with parents of a diverse pediatric cancer population before disclosure of WES results (n=64), then one to eight months (n=33) after disclosure. Interview transcripts were analyzed using a thematic qualitative approach. Results Parents identified a broad range of types of utility for their child's WES results. Even when results did not affect their child's current treatment, they expressed optimism about future clinical utility for their child, themselves, and other family members. Parents also reported experiencing psychological utility including peace of mind, relief of guilt, and satisfaction of curiosity. Pragmatic utility, such as the ability to plan for the future and make better reproductive decisions, was also described. Conclusion Parents of pediatric cancer patients perceive WES to have broad utility, including psychological and pragmatic utility, even if there is no direct impact on clinical care. Further work will need to consider how the value of genomic information should be characterized, how risks and benefits should be described, and how these results should inform recommendations and decisions about using WES.
Collapse
Affiliation(s)
- Janet Malek
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Melody J Slashinski
- School of Public Health & Health Sciences, University of Massachusetts, Amherst, MA
| | - Jill O Robinson
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Amanda M Gutierrez
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - D Williams Parsons
- Department of Pediatrics, Baylor College of Medicine, Houston, TX.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Sharon E Plon
- Department of Pediatrics, Baylor College of Medicine, Houston, TX.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Laurence B McCullough
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Amy L McGuire
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| |
Collapse
|
26
|
Kohler JN, Turbitt E, Biesecker BB. Personal utility in genomic testing: a systematic literature review. Eur J Hum Genet 2017; 25:662-668. [PMID: 28295040 DOI: 10.1038/ejhg.2017.10] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/20/2016] [Accepted: 01/11/2017] [Indexed: 11/09/2022] Open
Abstract
Researchers and clinicians refer to outcomes of genomic testing that extend beyond clinical utility as 'personal utility'. No systematic delineation of personal utility exists, making it challenging to appreciate its scope. Identifying empirical elements of personal utility reported in the literature offers an inventory that can be subsequently ranked for its relative value by those who have undergone genomic testing. A systematic review was conducted of the peer-reviewed literature reporting non-health-related outcomes of genomic testing from 1 January 2003 to 5 August 2016. Inclusion criteria specified English language, date of publication, and presence of empirical evidence. Identified outcomes were iteratively coded into unique domains. The search returned 551 abstracts from which 31 studies met the inclusion criteria. Study populations and type of genomic testing varied. Coding resulted in 15 distinct elements of personal utility, organized into three domains related to personal outcomes: affective, cognitive, and behavioral; and one domain related to social outcomes. The domains of personal utility may inform pre-test counseling by helping patients anticipate potential value of test results beyond clinical utility. Identified elements may also inform investigations into the prevalence and importance of personal utility to future test users.
Collapse
Affiliation(s)
- Jennefer N Kohler
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erin Turbitt
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Barbara B Biesecker
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Dias MM, Sorich MJ, Rowland A, Wiese MD, McKinnon RA. The Routine Clinical use of Pharmacogenetic Tests: What it Will Require? Pharm Res 2017; 34:1544-1550. [PMID: 28236061 DOI: 10.1007/s11095-017-2128-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/16/2017] [Indexed: 01/04/2023]
Abstract
Pharmacogenetic testing aims to personalize drug therapy with a view to optimising drug efficacy and minimise toxicity. However, despite the potential benefits, pharmacogenetic testing is mostly confined to specialised medical areas, laboratories and centres. Widespread integration into routine clinical practice has been limited by a complex set of issues including regulatory and reimbursement frameworks, evidence of clinical utility and clinician perspectives, practices and education. Here we assess the current barriers to widespread clinical uptake and identify the key issue necessary to address to accelerate routine testing.
Collapse
Affiliation(s)
- Mafalda M Dias
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia.,Department of Clinical Pharmacology, School of Medicine, Flinders University, Bedford Park, Australia
| | - Michael J Sorich
- Department of Clinical Pharmacology, School of Medicine, Flinders University, Bedford Park, Australia.,Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia
| | - Andrew Rowland
- Department of Clinical Pharmacology, School of Medicine, Flinders University, Bedford Park, Australia.,Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia
| | - Michael D Wiese
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia.
| |
Collapse
|
28
|
The value of genetic testing: beyond clinical utility. Genet Med 2016; 19:763-771. [DOI: 10.1038/gim.2016.186] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/06/2016] [Indexed: 12/26/2022] Open
|
29
|
Atwater T, Cook CM, Massion PP. The Pursuit of Noninvasive Diagnosis of Lung Cancer. Semin Respir Crit Care Med 2016; 37:670-680. [PMID: 27732989 DOI: 10.1055/s-0036-1592314] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The noninvasive diagnosis of lung cancer remains a formidable challenge. Although tissue diagnosis will remain the gold standard for the foreseeable future, questions pertaining to the risks and costs associated with invasive diagnostic procedures are of prime relevance. This review addresses new modalities for improving the noninvasive evaluation of suspicious lung nodules. Ultimately, the goal is to translate early diagnosis into early treatment. We discuss how biomarkers could assist in distinguishing benign from malignant nodules and aggressive from indolent tumors. The field of biomarkers is rapidly expanding and progressing, and efforts are well underway to apply molecular diagnostics to address the shortcomings of current lung cancer diagnostic tools.
Collapse
Affiliation(s)
- Thomas Atwater
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M Cook
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Pierre P Massion
- Cornelius Vanderbilt Endowed Chair in Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
30
|
Primary Care Providers' Interest in Using a Genetic Test to Guide Alcohol Use Disorder Treatment. J Subst Abuse Treat 2016; 70:14-20. [PMID: 27692183 DOI: 10.1016/j.jsat.2016.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/02/2016] [Accepted: 07/25/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Efforts to identify genetic moderators of pharmacotherapy response have generated interest in clinical applications of pharmacogenetic tests in alcohol use disorder (AUD) treatment. To date, no research on providers' interest in using pharmacogenetic tests in the context of AUD treatment has been reported. We conducted qualitative interviews with primary care providers from 5 clinics in the Veterans Health Administration (VA) to assess their interest in using a hypothetical genetic test to inform treatment of AUD with pharmacotherapy. METHODS Key contacts were used to recruit 24 providers from 5 primary care clinics associated with a single large VA medical facility. Participants completed 30-minute in-person semi-structured interviews focused on barriers and facilitators to provision of pharmacotherapy for AUD. Interviews included a hypothetical scenario regarding the availability of a genetic test to inform AUD pharmacotherapy provision and/or selection. Provider responses to the hypothetical scenario were recorded, transcribed and analyzed qualitatively using inductive content analysis. Data were independently coded by three investigators, and themes were identified via consensus. RESULTS Participants were generally interested in a genetic test to aid in AUD treatment planning. Five common themes were identified, including: perceived benefits of a pharmacogenetic test (e.g., aiding with therapeutic choice, positively impacting patient motivation for and engagement with AUD treatment), perceived drawbacks (e.g., limiting potential benefits of pharmacotherapy by reducing the target population for its receipt, adverse impacts of "negative" results), caveats to clinical utility (e.g., utility would depend on prognostic accuracy and/or medication characteristics), uncertainty as to whether such a test would impact clinical decision-making, and pragmatic barriers to use (costs and other resources, such as laboratory facilities). CONCLUSIONS Primary care providers in this study generally believed a genetic test to aid in AUD treatment planning would be useful, due to its potential to hone treatment choice as well as to influence patient motivation and adherence to treatment. However, providers acknowledged that a test's utility would depend on the strength of its prognostic characteristics, its other benefits relative to standard care, and lack of pragmatic barriers.
Collapse
|
31
|
Dotson WD, Bowen MS, Kolor K, Khoury MJ. Clinical utility of genetic and genomic services: context matters. Genet Med 2016; 18:672-4. [PMID: 26656648 PMCID: PMC4902786 DOI: 10.1038/gim.2015.153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/14/2015] [Indexed: 11/08/2022] Open
Affiliation(s)
- W. David Dotson
- 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
| | - 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
|
32
|
Garcia J, Tahiliani J, Johnson NM, Aguilar S, Beltran D, Daly A, Decker E, Haverfield E, Herrera B, Murillo L, Nykamp K, Topper S. Clinical Genetic Testing for the Cardiomyopathies and Arrhythmias: A Systematic Framework for Establishing Clinical Validity and Addressing Genotypic and Phenotypic Heterogeneity. Front Cardiovasc Med 2016; 3:20. [PMID: 27446933 PMCID: PMC4921949 DOI: 10.3389/fcvm.2016.00020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
Advances in DNA sequencing have made large, diagnostic gene panels affordable and efficient. Broad adoption of such panels has begun to deliver on the promises of personalized medicine, but has also brought new challenges such as the presence of unexpected results, or results of uncertain clinical significance. Genetic analysis of inherited cardiac conditions is particularly challenging due to the extensive genetic heterogeneity underlying cardiac phenotypes, and the overlapping, variable, and incompletely penetrant nature of their clinical presentations. The design of effective diagnostic tests and the effective use of the results depend on a clear understanding of the relationship between each gene and each considered condition. To address these issues, we developed simple, systematic approaches to three fundamental challenges: (1) evaluating the strength of the evidence suggesting that a particular condition is caused by pathogenic variants in a particular gene, (2) evaluating whether unusual genotype/phenotype observations represent a plausible expansion of clinical phenotype associated with a gene, and (3) establishing a molecular diagnostic strategy to capture overlapping clinical presentations. These approaches focus on the systematic evaluation of the pathogenicity of variants identified in clinically affected individuals, and the natural history of disease in those individuals. Here, we applied these approaches to the evaluation of more than 100 genes reported to be associated with inherited cardiomyopathies and arrhythmias including hypertrophic cardiomyopathy, dilated cardiomyopathy, arrhythmogenic right ventricular dysplasia or cardiomyopathy, long QT syndrome, short QT syndrome, Brugada, and catecholaminergic polymorphic ventricular tachycardia, and to a set of related syndromes such as Noonan Syndrome and Fabry disease. These approaches provide a framework for delivering meaningful and accurate genetic test results to individuals with hereditary cardiac conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Amy Daly
- Invitae Corporation, San Francisco, CA, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Harris LN, Ismaila N, McShane LM, Andre F, Collyar DE, Gonzalez-Angulo AM, Hammond EH, Kuderer NM, Liu MC, Mennel RG, Van Poznak C, Bast RC, Hayes DF. Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34:1134-50. [PMID: 26858339 PMCID: PMC4933134 DOI: 10.1200/jco.2015.65.2289] [Citation(s) in RCA: 592] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To provide recommendations on appropriate use of breast tumor biomarker assay results to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer. METHODS A literature search and prospectively defined study selection sought systematic reviews, meta-analyses, randomized controlled trials, prospective-retrospective studies, and prospective comparative observational studies published from 2006 through 2014. Outcomes of interest included overall survival and disease-free or recurrence-free survival. Expert panel members used informal consensus to develop evidence-based guideline recommendations. RESULTS The literature search identified 50 relevant studies. One randomized clinical trial and 18 prospective-retrospective studies were found to have evaluated the clinical utility, as defined by the guideline, of specific biomarkers for guiding decisions on the need for adjuvant systemic therapy. No studies that met guideline criteria for clinical utility were found to guide choice of specific treatments or regimens. RECOMMENDATIONS In addition to estrogen and progesterone receptors and human epidermal growth factor receptor 2, the panel found sufficient evidence of clinical utility for the biomarker assays Oncotype DX, EndoPredict, PAM50, Breast Cancer Index, and urokinase plasminogen activator and plasminogen activator inhibitor type 1 in specific subgroups of breast cancer. No biomarker except for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 was found to guide choices of specific treatment regimens. Treatment decisions should also consider disease stage, comorbidities, and patient preferences.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents, Hormonal/therapeutic use
- Biomarkers, Tumor/analysis
- Breast Neoplasms/chemistry
- Breast Neoplasms/drug therapy
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/chemistry
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Chemotherapy, Adjuvant
- Clinical Decision-Making/methods
- Comorbidity
- Disease-Free Survival
- Evidence-Based Medicine
- Female
- Humans
- Neoplasm Staging
- Plasminogen Activator Inhibitor 1/analysis
- Predictive Value of Tests
- Randomized Controlled Trials as Topic
- Receptor, ErbB-2/analysis
- Receptors, Estrogen/analysis
- Receptors, Progesterone/analysis
- Reproducibility of Results
- Survival Analysis
- Urokinase-Type Plasminogen Activator/analysis
Collapse
Affiliation(s)
- Lyndsay N Harris
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Nofisat Ismaila
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI.
| | - Lisa M McShane
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Fabrice Andre
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Deborah E Collyar
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Ana M Gonzalez-Angulo
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Elizabeth H Hammond
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Nicole M Kuderer
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Minetta C Liu
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Robert G Mennel
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Catherine Van Poznak
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Robert C Bast
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Daniel F Hayes
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| |
Collapse
|
34
|
Horvath AR, Bossuyt PMM, Sandberg S, John AS, Monaghan PJ, Verhagen-Kamerbeek WDJ, Lennartz L, Cobbaert CM, Ebert C, Lord SJ. Setting analytical performance specifications based on outcome studies - is it possible? Clin Chem Lab Med 2016; 53:841-8. [PMID: 25996384 DOI: 10.1515/cclm-2015-0214] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/10/2015] [Indexed: 11/15/2022]
Abstract
The 1st Strategic Conference of the European Federation of Clinical Chemistry and Laboratory Medicine proposed a simplified hierarchy for setting analytical performance specifications (APS). The top two levels of the 1999 Stockholm hierarchy, i.e., evaluation of the effect of analytical performance on clinical outcomes and clinical decisions have been proposed to be replaced by one outcome-based model. This model can be supported by: (1a) direct outcome studies; and (1b) indirect outcome studies investigating the impact of analytical performance of the test on clinical classifications or decisions and thereby on the probability of patient relevant clinical outcomes. This paper reviews the need for outcome-based specifications, the most relevant types of outcomes to be considered, and the challenges and limitations faced when setting outcome-based APS. The methods of Model 1a and b are discussed and examples are provided for how outcome data can be translated to APS using the linked evidence and simulation or decision analytic techniques. Outcome-based APS should primarily reflect the clinical needs of patients; should be tailored to the purpose, role and significance of the test in a well defined clinical pathway; and should be defined at a level that achieves net health benefit for patients at reasonable costs. Whilst it is acknowledged that direct evaluations are difficult and may not be possible for all measurands, all other forms of setting APS should be weighed against that standard, and regarded as approximations. Better definition of the relationship between the analytical performance of tests and health outcomes can be used to set analytical performance criteria that aim to improve the clinical and cost-effectiveness of laboratory tests.
Collapse
|
35
|
Was it worth it? Patients' perspectives on the perceived value of genomic-based individualized medicine. J Community Genet 2016; 7:145-52. [PMID: 26860291 DOI: 10.1007/s12687-016-0260-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/10/2016] [Indexed: 10/22/2022] Open
Abstract
The value of genomic sequencing is often understood in terms of its ability to affect diagnosis or treatment. In these terms, successes occur only in a minority of cases. This paper presents views from patients who had exome sequencing done clinically to explore how they perceive the utility of genomic medicine. The authors used semi-structured, qualitative interviews in order to study patients' attitudes toward genomic sequencing in oncology and rare-disease settings. Participants from 37 cases were interviewed. In terms of the testing's key values-regardless of having received what clinicians described as meaningful results-participants expressed four qualities that are separate from traditional views of clinical utility: Participants felt they had been empowered over their own health. They felt they had contributed altruistically to the progress of genomic technology in medicine. They felt their suffering had been legitimated. They also felt a sense of closure, having done everything they could. Patients expressed overwhelmingly positive attitudes toward sequencing. Their rationale was not solely based on the results' clinical utility. It is important for clinicians to understand this non-medical reasoning as it pertains to patient decision-making and informed consent.
Collapse
|
36
|
Khangura SD, Tingley K, Chakraborty P, Coyle D, Kronick JB, Laberge AM, Little J, Miller FA, Mitchell JJ, Prasad C, Siddiq S, Siriwardena K, Sparkes R, Speechley KN, Stockler S, Trakadis Y, Wilson BJ, Wilson K, Potter BK. Child and family experiences with inborn errors of metabolism: a qualitative interview study with representatives of patient groups. J Inherit Metab Dis 2016; 39. [PMID: 26209272 PMCID: PMC4710640 DOI: 10.1007/s10545-015-9881-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Patient-centered health care for children with inborn errors of metabolism (IEM) and their families is important and requires an understanding of patient experiences, needs, and priorities. IEM-specific patient groups have emerged as important voices within these rare disease communities and are uniquely positioned to contribute to this understanding. We conducted qualitative interviews with IEM patient group representatives to increase understanding of patient and family experiences, needs, and priorities and inform patient-centered research and care. METHODS We developed a sampling frame of patient groups representing IEM disease communities from Canada, the United States, and United Kingdom. With consent, we interviewed participants to explore their views on experiences, needs, and outcomes that are most important to children with IEM and their families. We analyzed the data using a qualitative descriptive approach to identify key themes and sub-themes. RESULTS We interviewed 18 organizational representatives between February 28 and September 17, 2014, representing 16 IEMs and/or disease categories. Twelve participants voluntarily self-identified as parents and/or were themselves patients. Three key themes emerged from the coded data: managing the uncertainty associated with raising and caring for a child with a rare disease; challenges associated with the affected child's life transitions, and; the collective struggle for improved outcomes and interventions that rare disease communities navigate. CONCLUSION Health care providers can support children with IEM and their families by acknowledging and reducing uncertainty, supporting families through children's life transitions, and contributing to rare disease communities' progress toward improved interventions, experiences, and outcomes.
Collapse
Affiliation(s)
- Sara D Khangura
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Kylie Tingley
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Pranesh Chakraborty
- Newborn Screening Ontario, Ottawa, ON, Canada
- Division of Metabolics and Newborn Screening, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Faculty of Medicine, Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Doug Coyle
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Jonathan B Kronick
- Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | | | - Julian Little
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Fiona A Miller
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | | | | | - Shabnaz Siddiq
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | | | | | | | | | | | - Brenda J Wilson
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | | | - Beth K Potter
- Faculty of Medicine, School of Public Health and Preventive Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
37
|
Van Poznak C, Somerfield MR, Bast RC, Cristofanilli M, Goetz MP, Gonzalez-Angulo AM, Hicks DG, Hill EG, Liu MC, Lucas W, Mayer IA, Mennel RG, Symmans WF, Hayes DF, Harris LN. Use of Biomarkers to Guide Decisions on Systemic Therapy for Women With Metastatic Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2015. [PMID: 26195705 DOI: 10.1200/jco.2015.61.1459] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To provide recommendations on the appropriate use of breast tumor biomarker assay results to guide decisions on systemic therapy for metastatic breast cancer. METHODS A literature search and prospectively defined study selection identified systematic reviews, meta-analyses, randomized controlled trials (RCTs), prospective-retrospective studies, and prospective comparative observational studies published from 2006 through September 2014. RESULTS The literature search revealed 17 articles that met criteria for further review: 11 studies reporting discordances between primary tumors and metastases in expression of hormone receptors or human epidermal growth factor receptor 2 (HER2), one RCT that addressed the use of a biomarker to decide whether to change or continue a treatment regimen, and five prospective-retrospective studies that evaluated the clinical utility of biomarkers. RECOMMENDATIONS In patients with accessible metastases, biopsy for confirmation of disease process and retesting of estrogen receptor, progesterone receptor, and HER2 status should be offered, but evidence is lacking to determine whether changing anticancer treatment on the basis of change in receptor status affects clinical outcomes. With discordance of results between primary and metastatic tissues, the Panel consensus is to use preferentially the estrogen receptor, progesterone receptor, and HER2 status of the metastasis to direct therapy if supported by the clinical scenario and patient's goals for care. Carcinoembryonic antigen, cancer antigen 15-3, and cancer antigen 27-29 may be used as adjunctive assessments, but not alone, to contribute to decisions regarding therapy. Recommendations for tumor rebiopsy and use of circulating tumor markers are based on clinical experience and Panel informal consensus in the absence of studies designed to evaluate the clinical utility of the markers. As such, it is also reasonable for clinicians to not use these markers as adjunctive assessments.
Collapse
Affiliation(s)
- Catherine Van Poznak
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Mark R Somerfield
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Robert C Bast
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Massimo Cristofanilli
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Matthew P Goetz
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Ana M Gonzalez-Angulo
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - David G Hicks
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Elizabeth G Hill
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Minetta C Liu
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Wanda Lucas
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Ingrid A Mayer
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Robert G Mennel
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - William F Symmans
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Daniel F Hayes
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Lyndsay N Harris
- Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Mark R. Somerfield, American Society of Clinical Oncology, Alexandria, VA; Robert C. Bast, Ana M. Gonzalez-Angulo, and William F. Symmans, University of Texas MD Anderson Cancer Center, Houston; Robert G. Mennel, Texas Oncology, Dallas, TX; Massimo Cristofanilli, Thomas Jefferson University-Kimmel Cancer Center, Philadelphia, PA; Matthew P. Goetz and Minetta C. Liu, Mayo Clinic, Rochester, MN; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Elizabeth G. Hill, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC; Wanda Lucas, Georgetown University, Washington, DC; Ingrid A. Mayer, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN; and Lyndsay N. Harris, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
38
|
Reichel E, Aldave AJ, Schaumberg DA, Singh R, Henderson BA. Genetic testing for age-related macular degeneration: progress and perspectives. EXPERT REVIEW OF OPHTHALMOLOGY 2015. [DOI: 10.1586/17469899.2015.1059752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Zota VE, Magliocco AM. Molecular Technologies in the Clinical Diagnostic Laboratory. Cancer Control 2015; 22:142-51. [PMID: 26068758 DOI: 10.1177/107327481502200204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND New technologies for molecular analysis are increasing our ability to diagnose cancer. METHODS Several molecular analysis technologies are reviewed and their use in the clinical laboratory is discussed. RESULTS Select key technologies, including polymerase chain reaction and next-generation sequencing, are helping transform our ability to analyze cancer specimens. As these technological advances become more and more incorporated into routine diagnostic testing, our classification systems are likely to be impacted and our approach to treatment transformed. The routine use of such technology also brings challenges for analysis and reimbursement. CONCLUSION These advances in technology will change the way we diagnose, monitor, and treat patients with cancer.
Collapse
Affiliation(s)
- Victor E Zota
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | |
Collapse
|
40
|
Vachani A, Pass HI, Rom WN, Midthun DE, Edell ES, Laviolette M, Li XJ, Fong PY, Hunsucker SW, Hayward C, Mazzone PJ, Madtes DK, Miller YE, Walker MG, Shi J, Kearney P, Fang KC, Massion PP. Validation of a multiprotein plasma classifier to identify benign lung nodules. J Thorac Oncol 2015; 10:629-37. [PMID: 25590604 PMCID: PMC4382127 DOI: 10.1097/jto.0000000000000447] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Indeterminate pulmonary nodules (IPNs) lack clinical or radiographic features of benign etiologies and often undergo invasive procedures unnecessarily, suggesting potential roles for diagnostic adjuncts using molecular biomarkers. The primary objective was to validate a multivariate classifier that identifies likely benign lung nodules by assaying plasma protein expression levels, yielding a range of probability estimates based on high negative predictive values (NPVs) for patients with 8 to 30 mm IPNs. METHODS A retrospective, multicenter, case-control study was performed using multiple reaction monitoring mass spectrometry, a classifier comprising five diagnostic and six normalization proteins, and blinded analysis of an independent validation set of plasma samples. RESULTS The classifier achieved validation on 141 lung nodule-associated plasma samples based on predefined statistical goals to optimize sensitivity. Using a population based nonsmall-cell lung cancer prevalence estimate of 23% for 8 to 30 mm IPNs, the classifier identified likely benign lung nodules with 90% negative predictive value and 26% positive predictive value, as shown in our prior work, at 92% sensitivity and 20% specificity, with the lower bound of the classifier's performance at 70% sensitivity and 48% specificity. Classifier scores for the overall cohort were statistically independent of patient age, tobacco use, nodule size, and chronic obstructive pulmonary disease diagnosis. The classifier also demonstrated incremental diagnostic performance in combination with a four-parameter clinical model. CONCLUSIONS This proteomic classifier provides a range of probability estimates for the likelihood of a benign etiology that may serve as a noninvasive, diagnostic adjunct for clinical assessments of patients with IPNs.
Collapse
Affiliation(s)
- Anil Vachani
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Harvey I. Pass
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - William N. Rom
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - David E. Midthun
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Eric S. Edell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Michel Laviolette
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Xiao-Jun Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Pui-Yee Fong
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Stephen W. Hunsucker
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Clive Hayward
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Peter J. Mazzone
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - David K. Madtes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - York E. Miller
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Michael G. Walker
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Jing Shi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Paul Kearney
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Kenneth C. Fang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| | - Pierre P. Massion
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn Lung Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and Department of Environmental Medicine, New York University School of Medicine, New York University Langone Medical Center, New York, New York; Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota; Unité de Recherche en Pneumologie, Centre de Recherche de l’Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumologie de l’Université Laval; Québec, Canada; Integrated Diagnostics, Seattle, Washington; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Veterans Administration Eastern Colorado Healthcare System, University of Colorado Denver School of Medicine, Denver, Colorado; Statistics Consultant, Carlsbad, California; Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee
| |
Collapse
|
41
|
Bowdin SC, Hayeems RZ, Monfared N, Cohn RD, Meyn MS. The SickKids Genome Clinic: developing and evaluating a pediatric model for individualized genomic medicine. Clin Genet 2015; 89:10-9. [PMID: 25813238 DOI: 10.1111/cge.12579] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 02/01/2015] [Accepted: 02/23/2015] [Indexed: 01/16/2023]
Abstract
Our increasing knowledge of how genomic variants affect human health and the falling costs of whole-genome sequencing are driving the development of individualized genomic medicine. This new clinical paradigm uses knowledge of an individual's genomic variants to anticipate, diagnose and manage disease. While individualized genetic medicine offers the promise of transformative change in health care, it forces us to reconsider existing ethical, scientific and clinical paradigms. The potential benefits of pre-symptomatic identification of at-risk individuals, improved diagnostics, individualized therapy, accurate prognosis and avoidance of adverse drug reactions coexist with the potential risks of uninterpretable results, psychological harm, outmoded counseling models and increased health care costs. Here we review the challenges, opportunities and limits of integrating genomic analysis into pediatric clinical practice and describe a model for implementing individualized genomic medicine. Our multidisciplinary team of bioinformaticians, health economists, health services and policy researchers, ethicists, geneticists, genetic counselors and clinicians has designed a 'Genome Clinic' research project that addresses multiple challenges in pediatric genomic medicine--ranging from development of bioinformatics tools for the clinical assessment of genomic variants and the discovery of disease genes to health policy inquiries, assessment of clinical care models, patient preference and the ethics of consent.
Collapse
Affiliation(s)
- S C Bowdin
- Division of Clinical and Metabolic Genetics, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada.,Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, Canada.,Department of Paediatrics, University of Toronto, Toronto, Canada
| | - R Z Hayeems
- Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, Canada.,Program in Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Canada.,Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Canada
| | - N Monfared
- Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, Canada
| | - R D Cohn
- Division of Clinical and Metabolic Genetics, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada.,Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Paediatrics, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - M S Meyn
- Division of Clinical and Metabolic Genetics, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada.,Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Paediatrics, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
42
|
Parkinson DR, McCormack RT, Keating SM, Gutman SI, Hamilton SR, Mansfield EA, Piper MA, Deverka P, Frueh FW, Jessup JM, McShane LM, Tunis SR, Sigman CC, Kelloff GJ. Evidence of clinical utility: an unmet need in molecular diagnostics for patients with cancer. Clin Cancer Res 2014; 20:1428-44. [PMID: 24634466 DOI: 10.1158/1078-0432.ccr-13-2961] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article defines and describes best practices for the academic and business community to generate evidence of clinical utility for cancer molecular diagnostic assays. Beyond analytical and clinical validation, successful demonstration of clinical utility involves developing sufficient evidence to demonstrate that a diagnostic test results in an improvement in patient outcomes. This discussion is complementary to theoretical frameworks described in previously published guidance and literature reports by the U.S. Food and Drug Administration, Centers for Disease Control and Prevention, Institute of Medicine, and Center for Medical Technology Policy, among others. These reports are comprehensive and specifically clarify appropriate clinical use, adoption, and payer reimbursement for assay manufacturers, as well as Clinical Laboratory Improvement Amendments-certified laboratories, including those that develop assays (laboratory developed tests). Practical criteria and steps for establishing clinical utility are crucial to subsequent decisions for reimbursement without which high-performing molecular diagnostics will have limited availability to patients with cancer and fail to translate scientific advances into high-quality and cost-effective cancer care. See all articles in this CCR Focus section, "The Precision Medicine Conundrum: Approaches to Companion Diagnostic Co-development."
Collapse
Affiliation(s)
- David R Parkinson
- Authors' Affiliations: New Enterprise Associates, Inc., Menlo Park; CCS Associates, Mountain View; Myraqa, Redwood Shores, California; Johnson & Johnson/Veridex, LLC, Raritan, New Jersey; University of Texas, MD Anderson Cancer Center, Houston, Texas; Center for Diagnostics and Radiologic Health, Office of In Vitro Diagnostics, Personalized Medicine Program, Silver Spring; Center for Medical Technology Policy, Baltimore; Opus Three LLC; National Cancer Institute, Division of Cancer Treatment and Diagnosis, Rockville, Maryland; and Kaiser Permanente Research Affiliates Evidence-Based Practice Center, Kaiser Permanente Center for Health Research, Portland, Oregon
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
|
45
|
Hamilton JG, Edwards HM, Khoury MJ, Taplin SH. Cancer screening and genetics: a tale of two paradigms. Cancer Epidemiol Biomarkers Prev 2014; 23:909-16. [PMID: 24706727 PMCID: PMC4047129 DOI: 10.1158/1055-9965.epi-13-1016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The long-standing medical tradition to "first do no harm" is reflected in population-wide evidence-based recommendations for cancer screening tests that focus primarily on reducing morbidity and mortality. The conventional cancer screening process is predicated on finding early-stage disease that can be treated effectively; yet emerging genetic and genomic testing technologies have moved the target earlier in the disease development process to identify a probabilistic predisposition to disease. Genetic risk information can have varying implications for the health and well-being of patients and their relatives, and has raised important questions about the evaluation and value of risk information. This article explores the paradigms that are being applied to the evaluation of conventional cancer screening tests and emerging genetic and genomic tests of cancer susceptibility, and how these perspectives are shifting and evolving in response to advances in our ability to detect cancer risks. We consider several challenges germane to the evaluation of both categories of tests, including defining benefits and harms in terms of personal and clinical utility, addressing healthcare consumers' information preferences, and managing scientific uncertainty. We encourage research and dialogue aimed at developing a better understanding of the value of all risk information, nongenetic and genetic, to people's lives. Cancer Epidemiol Biomarkers Prev; 23(6); 909-16. ©2014 AACR.
Collapse
Affiliation(s)
- Jada G Hamilton
- Authors' Affiliations: Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York; Clinical Research Directorate/CMRP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick; Epidemiology and Genomics Research Program, Process of Care Research Branch, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland; and Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Heather M Edwards
- Authors' Affiliations: Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York; Clinical Research Directorate/CMRP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick; Epidemiology and Genomics Research Program, Process of Care Research Branch, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland; and Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Muin J Khoury
- Authors' Affiliations: Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York; Clinical Research Directorate/CMRP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick; Epidemiology and Genomics Research Program, Process of Care Research Branch, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland; and Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, GeorgiaAuthors' Affiliations: Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York; Clinical Research Directorate/CMRP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick; Epidemiology and Genomics Research Program, Process of Care Research Branch, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland; and Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Stephen H Taplin
- Authors' Affiliations: Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York; Clinical Research Directorate/CMRP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick; Epidemiology and Genomics Research Program, Process of Care Research Branch, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland; and Office of Public Health Genomics, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
46
|
Bowdin S, Ray PN, Cohn RD, Meyn MS. The Genome Clinic: A Multidisciplinary Approach to Assessing the Opportunities and Challenges of Integrating Genomic Analysis into Clinical Care. Hum Mutat 2014; 35:513-9. [DOI: 10.1002/humu.22536] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/21/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Sarah Bowdin
- Division of Clinical and Metabolic Genetics; Department of Paediatrics; The Hospital for Sick Children; Toronto Ontario Canada
- The Centre for Genetic Medicine; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Paediatrics; University of Toronto; Toronto Ontario Canada
| | - Peter N. Ray
- The Centre for Genetic Medicine; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Paediatric Laboratory Medicine; The Hospital for Sick Children; Toronto Ontario Canada
- Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - Ronald D. Cohn
- Division of Clinical and Metabolic Genetics; Department of Paediatrics; The Hospital for Sick Children; Toronto Ontario Canada
- The Centre for Genetic Medicine; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Paediatrics; University of Toronto; Toronto Ontario Canada
- Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - M. Stephen Meyn
- Division of Clinical and Metabolic Genetics; Department of Paediatrics; The Hospital for Sick Children; Toronto Ontario Canada
- The Centre for Genetic Medicine; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Paediatrics; University of Toronto; Toronto Ontario Canada
- Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Molecular Genetics; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
47
|
McCarthy JJ, McLeod HL, Ginsburg GS. Genomic medicine: a decade of successes, challenges, and opportunities. Sci Transl Med 2014; 5:189sr4. [PMID: 23761042 DOI: 10.1126/scitranslmed.3005785] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Genomic medicine--an aspirational term 10 years ago--is gaining momentum across the entire clinical continuum from risk assessment in healthy individuals to genome-guided treatment in patients with complex diseases. We review the latest achievements in genome research and their impact on medicine, primarily in the past decade. In most cases, genomic medicine tools remain in the realm of research, but some tools are crossing over into clinical application, where they have the potential to markedly alter the clinical care of patients. In this State of the Art Review, we highlight notable examples including the use of next-generation sequencing in cancer pharmacogenomics, in the diagnosis of rare disorders, and in the tracking of infectious disease outbreaks. We also discuss progress in dissecting the molecular basis of common diseases, the role of the host microbiome, the identification of drug response biomarkers, and the repurposing of drugs. The significant challenges of implementing genomic medicine are examined, along with the innovative solutions being sought. These challenges include the difficulty in establishing clinical validity and utility of tests, how to increase awareness and promote their uptake by clinicians, a changing regulatory and coverage landscape, the need for education, and addressing the ethical aspects of genomics for patients and society. Finally, we consider the future of genomics in medicine and offer a glimpse of the forces shaping genomic medicine, such as fundamental shifts in how we define disease, how medicine is delivered to patients, and how consumers are managing their own health and affecting change.
Collapse
Affiliation(s)
- Jeanette J McCarthy
- Institute for Genome Sciences & Policy, Duke University, Durham, NC 27708, USA
| | | | | |
Collapse
|
48
|
Goddard KAB, Whitlock EP, Berg JS, Williams MS, Webber EM, Webster JA, Lin JS, Schrader KA, Campos-Outcalt D, Offit K, Feigelson HS, Hollombe C. Description and pilot results from a novel method for evaluating return of incidental findings from next-generation sequencing technologies. Genet Med 2013; 15:721-8. [PMID: 23558254 PMCID: PMC3927794 DOI: 10.1038/gim.2013.37] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 02/25/2013] [Indexed: 12/23/2022] Open
Abstract
PURPOSE The aim of this study was to develop, operationalize, and pilot test a transparent, reproducible, and evidence-informed method to determine when to report incidental findings from next-generation sequencing technologies. METHODS Using evidence-based principles, we proposed a three-stage process. Stage I "rules out" incidental findings below a minimal threshold of evidence and is evaluated using inter-rater agreement and comparison with an expert-based approach. Stage II documents criteria for clinical actionability using a standardized approach to allow experts to consistently consider and recommend whether results should be routinely reported (stage III). We used expert opinion to determine the face validity of stages II and III using three case studies. We evaluated the time and effort for stages I and II. RESULTS For stage I, we assessed 99 conditions and found high inter-rater agreement (89%), and strong agreement with a separate expert-based method. Case studies for familial adenomatous polyposis, hereditary hemochromatosis, and α1-antitrypsin deficiency were all recommended for routine reporting as incidental findings. The method requires <3 days per topic. CONCLUSION We establish an operational definition of clinically actionable incidental findings and provide documentation and pilot testing of a feasible method that is scalable to the whole genome.
Collapse
Affiliation(s)
- Katrina A B Goddard
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
The EGAPP initiative: lessons learned. Genet Med 2013; 16:217-24. [PMID: 23928914 DOI: 10.1038/gim.2013.110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/24/2013] [Indexed: 01/12/2023] Open
Abstract
The Evaluation of Genomic Applications in Practice and Prevention Working Group was first convened in 2005 to develop and test evidence-based methods for the evaluation of genomic tests in transition from research to clinical and public health practice. Over the ensuing years, the Working Group has met 26 times, publishing eight recommendation statements, two methods papers, and one outcomes paper, as well as planning and serving as technical experts on numerous associated systematic reviews. Evaluation of Genomic Applications in Practice and Prevention methods have evolved to address implications of the proliferation of genome-wide association studies and are currently expanding to face challenges expected from clinical implementation of whole-genome sequencing tests. In this article, we review the work of the Evaluation of Genomic Applications in Practice and Prevention Working Group over the first 8 years of its existence with an emphasis on lessons learned throughout the process. It is hoped that in addition to the published methods of the Working Group, the lessons we have learned along the way will be informative to others who are producers and consumers of evidence-based guidelines in the field of genomic medicine.
Collapse
|
50
|
Fears R, ter Meulen V, Participants in the Working Group were: Stefania Boccia, Martina Cornel, Marc Delpech, Anne De Paepe, Xavier Estivill, Mats Hansson, Katia Karalis, Andres Metspalu, Markus Nöthen, Peter Propping, Jorge Sequeiros, Ron Zimmern, Volker ter Meulen (Chair), Robin Fears (secretariat). The perspective from EASAC and FEAM on direct-to-consumer genetic testing for health-related purposes. Eur J Hum Genet 2013; 21:703-7. [PMID: 23169492 PMCID: PMC3722941 DOI: 10.1038/ejhg.2012.238] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Direct-to-consumer (DTC) genetic testing services raise scientific, regulatory and ethical questions. A report was prepared by consultation with an expert Working Group and published by the academies of science (European Academies of Science Advisory Council, EASAC) and medicine (Federation of European Academies of Medicine, FEAM). This report reviews current scientific evidence, ascertains the principles that should underpin the options for action by policy-makers, and discusses the potential for devising proportionate and flexible regulation that enables future innovation, taking account of the work of other expert groups, most notably the European Society of Human Genetics. EASAC-FEAM concluded that DTC genetic testing has little clinical value at present, and expresses especial caution in several specific respects, for example relating to testing for high penetrance, serious disorders, prenatal screening, nutrigenomic and pharmacogenetic testing. It was emphasised that regulation must be on the basis that claims about the link between genetic marker and disease are scientifically valid. Other key issues to address include quality assurance (that includes the professional interpretation of results), transparent supply of accurate information, consideration of the implications for established health services, and clarification of consent procedures for any use of data for research purposes. There are important implications: for the European Commission, in revising the Directive on In Vitro Diagnostic Medical Devices; for professional bodies, in supporting training and guideline development; for the broader research community, in generating the evidence base; and for the public health community, in improving the routine translation of research advances into clinical practice.
Collapse
Affiliation(s)
- Robin Fears
- EASAC, German National Academy of Sciences Leopoldina, Saale, Germany
| | - Volker ter Meulen
- EASAC, German National Academy of Sciences Leopoldina, Saale, Germany
| | - Participants in the Working Group were: Stefania Boccia, Martina Cornel, Marc Delpech, Anne De Paepe, Xavier Estivill, Mats Hansson, Katia Karalis, Andres Metspalu, Markus Nöthen, Peter Propping, Jorge Sequeiros, Ron Zimmern, Volker ter Meulen (Chair), Robin Fears (secretariat)
- EASAC, German National Academy of Sciences Leopoldina, Saale, Germany
| |
Collapse
|