1
|
Flowers-Moore T, Rapp AR, Salazar JH, Rajendran R. Evaluation of pharmacogenomic testing to identify cytochrome P450 and SLCO1B1 enzymes and adverse drug events: A non-experimental observational research. Medicine (Baltimore) 2025; 104:e42031. [PMID: 40193664 PMCID: PMC11977719 DOI: 10.1097/md.0000000000042031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 02/18/2025] [Accepted: 03/16/2025] [Indexed: 04/09/2025] Open
Abstract
A laboratory-initiated preemptive and reactive cytochrome P450 and SLCO1B1 PGx testing protocol was evaluated in a private toxicology laboratory with the intent of identifying enzyme frequencies and associated adverse drug events. This study involved non-experimental observational research. During the retrospective medical chart review, patient demographics, statements of medical necessity, and PGx testing data were collected. Frequencies and percentages were calculated for the collected data, and statistical analysis was performed using Intellectus online software. A total of 192 PGx patient records from September 2019 to October 2021 were retrospectively reviewed. For patient demographics, men (n = 118; (61%)) were the majority gender identified among the patient population and Caucasians (n = 112; (58%)) followed by African Americans (n = 37; (19%)) were the most identified ancestry. The mean age of the patients was 69 (±9) years. CYP1A2 hyperinducers, followed by CYP3A5 poor metabolizers and CYP2B6 intermediate metabolizers, are the most encountered cytochrome P450 and SLCO1B1 enzymes. Regarding drug-gene interactions, 41 patients had 1 interaction, 29 had 2, and 31 had 3 or more interactions. For drug-drug interactions, 35 patients had 1 interaction, 15 had 2, and 30 had 3 or more interactions. Overall, 123 patients showed a minor or greater impact on drug-drug or drug-gene interactions. Overall, our study identified cytochrome P450 and SCLCO1B1 enzyme frequencies and patients experiencing actionable adverse drug events. By raising awareness of PGx test results through individualized clinician training, education, and interventions, these adverse events can be promptly identified and resolved.
Collapse
Affiliation(s)
- Tiffany Flowers-Moore
- Department of Clinical Laboratory Sciences, University of Texas Medical Branch, Galveston, TX
| | - Alexandra R. Rapp
- Department of Clinical Laboratory Sciences, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| | - Jose H. Salazar
- Department of Clinical Laboratory Sciences, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| | - Rajkumar Rajendran
- Department of Clinical Laboratory Sciences, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
2
|
Austin CA, Seligman B, Shan-Bala S, Kuchel GA, Loh KP, Kistler C, Batsis JA. Aging precisely: Precision medicine through the lens of an older adult. J Am Geriatr Soc 2024; 72:2972-2980. [PMID: 38888213 PMCID: PMC11461112 DOI: 10.1111/jgs.19036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024]
Abstract
Precision medicine presents an opportunity to use novel, data-driven strategies to improve patient care. The field of precision medicine has undergone many advancements over the past few years. It has moved beyond incorporation of individualized genetic risk into medical decision-making to include multiple other factors such as unique social, demographic, behavioral, and clinical characteristics. Geriatric medicine stands to benefit heavily from the integration of precision medicine into its standard practices. Older adults, compared with other populations, have high clinical and biological heterogeneity that can alter the risks and benefits of different approaches to patient care. These factors have not been routinely considered previously by geriatricians. Yet, geriatricians' ability to address older adults' baseline heterogeneity is increasingly recognized as a cornerstone of delivering quality care in a geriatric medical practice. Given the shared focus of individualized decision-making, precision medicine is a natural fit for geriatric medicine. This manuscript provides, via cases and discussion, examples that illustrate how precision medicine can improve the care of our older patients today. We will share specific and existing tools and evidence, and review the existing multilevel barriers to further incorporate and implement these tools into clinical practice. We propose methods to address these barriers and to help realize the full potential of precision medicine for the care of older adults. We conclude with a brief discussion of potential future directions of research of precision medicine in the care of older adults.
Collapse
Affiliation(s)
- C. Adrian Austin
- Division of Pulmonary and Critical Care Medicine, University of North Carolina, Chapel Hill, NC
- Division of Geriatric Medicine and Center for Aging and Health, University of North Carolina, Chapel Hill, NC
| | - Benjamin Seligman
- Geriatric Research, Education and Clinical Center, VA Greater Los Angeles Health Care System, Los Angeles, CA
- Division of Geriatric Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Sangeetha Shan-Bala
- Division of Geriatric Medicine, Department of Medicine, Inova Health System, Fairfax Medical Campus, Falls Church, VA
| | - George A. Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT
| | - Kah Poh Loh
- Division of Hematology/Oncology, Department of Medicine, James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Chrissy Kistler
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - John A. Batsis
- Division of Geriatric Medicine and Center for Aging and Health, University of North Carolina, Chapel Hill, NC
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
3
|
Choufani M, Kay J, Ermann J. Axial spondyloarthritis guidelines - aiming for maximum impact. Curr Opin Rheumatol 2024; 36:251-260. [PMID: 38661436 DOI: 10.1097/bor.0000000000001020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
PURPOSE OF REVIEW This review discusses international clinical practice guidelines (CPGs) for axial spondyloarthritis (axSpA) focusing on methodology, guideline quality, and implementation. RECENT FINDINGS The Assessment of SpondyloArthritis International Society/European Alliance of Associations for Rheumatology (ASAS/EULAR) and Pan-American League of Associations for Rheumatology (PANLAR) recently published axSpA CPGs and updates of the American College of Rheumatology/Spondylitis Association of America/Spondyloarthritis Research and Treatment Network (ACR/SAA/SPARTAN) and Asia-Pacific League of Associations for Rheumatology (APLAR) CPGs are expected. GRADE has emerged as the dominant framework for CPG development and has been used by three of the four international axSpA guidelines. Notable differences exist among these guidelines in the way that the recommendations are presented. Two of the four acknowledge the need for implementation strategies, but little detail about this is provided. The few studies that have evaluated the implementation of axSpA CPGs have identified poor adherence to recommendations on physical therapy/exercise and disease activity monitoring. Implementation science has identified many barriers and facilitators affecting guideline uptake, including those related to healthcare professionals and to the guidelines themselves. Creation of a tailored implementation plan simultaneously with the CPG is recommended. SUMMARY While methodological rigor in the creation of evidence-based recommendations is the focus of CPG development, recommendations must be presented in a user-friendly format that makes them easy to apply. 'Living guidelines' could facilitate keeping content up to date. Implementation is critical for the success of a CPG and should be emphasized in future axSpA guideline updates. Further research is needed to better understand the factors impacting the successful implementation of axSpA CPGs.
Collapse
Affiliation(s)
| | - Jonathan Kay
- UMass Chan Medical School and UMass Memorial Medical Center, Worcester
| | - Joerg Ermann
- Brigham and Women's Hospital, Boston
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Banerjee R, Cicero KI, Lee SS, Cowan AJ. Definers and drivers of functional high-risk multiple myeloma: insights from genomic, transcriptomic, and immune profiling. Front Oncol 2023; 13:1240966. [PMID: 37849816 PMCID: PMC10577204 DOI: 10.3389/fonc.2023.1240966] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Traditional prognostic models for newly diagnosed patients with multiple myeloma (MM), including International Staging System criteria and number of high-risk chromosomal abnormalities, are based on disease characteristics at diagnosis. However, the identification of patients at risk of more rapidly progressive MM is inherently a dynamic assessment. In a subset of patients with MM, adverse disease biology only becomes evident after the failure of first-line therapy. We define this entity as functional high-risk MM (FHRMM), encompassing relapse within 18 months of treatment initiation and/or within 12 months of frontline autologous stem cell transplantation. FHRMM is not adequately captured by traditional prognostic models, and there is a need for better understanding of mechanisms or risk factors for early relapse or progression. In this review, we explore potential definitions of FHRMM before delving into its underlying drivers based on genetic, transcriptomic, and immune cell profiling studies. Emerging data suggest that specific features of both myeloma cells and immune cells can enable the FHRMM phenotype. We conclude our review by discussing ongoing and future studies that seek to identify and intervene upon patients with FHRMM preemptively.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Kara I. Cicero
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sarah S. Lee
- Division of Myeloma, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, CA, United States
| | - Andrew J. Cowan
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
5
|
Lemke LK, Alam B, Williams R, Starostik P, Cavallari LH, Cicali EJ, Wiisanen K. Reimbursement of pharmacogenetic tests at a tertiary academic medical center in the United States. Front Pharmacol 2023; 14:1179364. [PMID: 37645439 PMCID: PMC10461057 DOI: 10.3389/fphar.2023.1179364] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/17/2023] [Indexed: 08/31/2023] Open
Abstract
Introduction: Pharmacogenetics (PGx) has the potential to improve health outcomes but cost of testing is a barrier for equitable access. Reimbursement by insurance providers may lessen the financial burden for patients, but the extent to which PGx claims are covered in clinical practice has not been well-characterized in the literature. Methods: A retrospective analysis of outpatient claims submitted to payers for PGx tests from 1/1/2019 through 12/31/2021 was performed. A reimbursement rate was calculated and compared across specific test types (e.g., single genes, panel), payers, indication, and the year the claim was submitted. Results: A total of 1,039 outpatient claims for PGx testing were analyzed. The overall reimbursement rate was 46% and ranged from 36%-48% across payers. PGx panels were reimbursed at a significantly higher rate than single gene tests (74% vs. 43%, p < 0.001). Discussion: Reimbursement of claims for PGx testing is variable based on the test type, indication, year the claim was submitted, number of diagnosis codes submitted, and number of unique diagnosis codes submitted. Due to the highly variable nature of reimbursement, cost and affordability should be discussed with each patient.
Collapse
Affiliation(s)
- Lauren K. Lemke
- Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Benish Alam
- Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Roy Williams
- Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
| | - Petr Starostik
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- UF Health Pathology Laboratories, UF Health, Gainesville, FL, United States
| | - Larisa H. Cavallari
- Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Emily J. Cicali
- Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Kristin Wiisanen
- Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
6
|
Fragala MS, Shaman JA, Lorenz RA, Goldberg SE. Role of Pharmacogenomics in Comprehensive Medication Management: Considerations for Employers. Popul Health Manag 2022; 25:753-762. [PMID: 36301527 DOI: 10.1089/pop.2022.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rising prescription costs, poor medication adherence, and safety issues pose persistent challenges to employer-sponsored health care plans and their beneficiaries. Comprehensive medication management (CMM), a patient-centered approach to medication optimization, enriched by pharmacogenomics (PGx), has been shown to improve the efficacy and safety of pharmaceutical regimens. This has contributed to improved health care outcomes, reduced costs of treatments, better adherence, shorter durations of treatment, and fewer adverse effects from drug therapy. Despite compelling clinical and economic evidence to justify the application of CMM guided by PGx, implementation in clinical settings remains sparse; notable barriers include limited physician adoption and health insurance coverage. Ultimately, these challenges may be overcome through comprehensive programs that include clinical decision support systems and education through employer-sponsored population health management channels to the benefit of the employees, employers, health care providers, and health care systems. This article discusses benefits, considerations, and barriers of scalable PGx-enriched CMM programs in the context of self-insured employers.
Collapse
|
7
|
Mittal S, Tang I, Gleeson JG. Evaluating human mutation databases for “treatability” using patient-customized therapy. MED 2022; 3:740-759. [DOI: 10.1016/j.medj.2022.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022]
|
8
|
Association between Health Insurance Type and Genetic Testing and/or Counseling for Breast and Ovarian Cancer. J Pers Med 2022; 12:jpm12081263. [PMID: 36013212 PMCID: PMC9409681 DOI: 10.3390/jpm12081263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
As genetic testing becomes increasingly incorporated into clinical practice to aid in both the diagnosis and risk assessment of genetic diseases, patients benefit from genetic counseling to support their understanding of test results either before and/or after genetic testing. Therefore, access to genetic testing and counseling is imperative for patient care. It is well established that health insurance coverage is a major determinant of access to health care in the United States as individuals without insurance are less likely to have a regular source of health care than their insured counterparts. Different health insurance plans and benefits also influence patients’ access to health care. Data on the association of health insurance and the uptake of genetic testing and/or counseling for cancer risk are limited. Using data from the National Health Interview Survey, we examined the uptake of genetic testing and/or counseling for breast/ovarian cancer risk by health insurance type. We found that only a small proportion of women undergo genetic testing and/or counseling for breast/ovarian cancer risk (2.3%), even among subgroups of women at risk due to family or personal history (6.5%). Women with health insurance were more likely to undergo genetic testing and/or counseling for breast/ovarian cancer risk, particularly those with military and private insurance plans, than those without health insurance after adjusting for various demographic, socioeconomic, and health risk covariates. Further investigations are needed to examine potential disparities in access and health inequities.
Collapse
|
9
|
Mansur A, Zhang F, Lu CY. Genetic Testing and/or Counseling for Colorectal Cancer by Health Insurance Type. J Pers Med 2022; 12:jpm12071146. [PMID: 35887643 PMCID: PMC9317363 DOI: 10.3390/jpm12071146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 12/18/2022] Open
Abstract
Genetic testing is increasingly used in clinical practice to assist with the diagnosis of genetic diseases and/or provide information about disease risk, and genetic counseling supports patient understanding of test results before and/or after genetic testing. Therefore, access to genetic testing and counseling is important for patient care. Health insurance coverage is a major determinant of access to health care in the United States. Uninsured individuals are less likely to have a regular source of health care than their insured counterparts. Different health insurance types and benefits also influence access to health care. Data on the association of health insurance and uptake of genetic testing and/or counseling for cancer risk are limited. Using data from the National Health Interview Survey, we examined the uptake of genetic testing and/or counseling for colorectal cancer (CRC) risk by health insurance type. We found that only a small proportion of individuals undergo genetic testing and/or counseling for CRC risk (0.8%), even among subgroups of individuals at risk due to family or personal history (3.7%). Insured individuals were more likely to undergo genetic testing and/or counseling for CRC risk, particularly those with Military and Private insurance plans, after adjusting for various demographic, socioeconomic, and health risk covariates. Further investigations are warranted to examine potential disparities in access and health inequities.
Collapse
Affiliation(s)
| | - Fang Zhang
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA 02215, USA;
| | - Christine Y. Lu
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA 02215, USA;
- Correspondence: ; Tel.: +1-617-867-4989
| |
Collapse
|
10
|
Holloway K, Miller FA. The Consultant's intermediary role in the regulation of molecular diagnostics in the US. Soc Sci Med 2022; 304:112929. [PMID: 32201019 DOI: 10.1016/j.socscimed.2020.112929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/28/2020] [Accepted: 03/12/2020] [Indexed: 11/17/2022]
Abstract
Molecular diagnostics are fast becoming a big business, with the promise of personalized medicine fueling the growth of "blockbuster" tests with high expectations for health system impact and commercial success. We investigate the polycentric regulatory regime for molecular diagnostics in the US, drawing attention to the prominent role of coverage and reimbursement systems in setting regulatory standards for this industry. We hone in on the private consultants who assist molecular diagnostics companies to gain broad clinical uptake of their products. Through a web-based search of consulting companies, analysis of their online materials, and 13 qualitative interviews with consultants, we describe the role of these actors in the coverage and reimbursement of novel diagnostics and highlight the production of evidence as a critical part of the process. We argue that consultants operate as regulatory intermediaries, helping to develop the evidentiary standards for payment decisions that ultimately benefit their clients, the manufacturers. We suggest that public policy discussions over how best to realize the promise of personalized medicine should be re-oriented to consider whose interests are represented in the regulatory regime governing access to these technologies.
Collapse
|
11
|
Anderson HD, Thant TM, Kao DP, Crooks KR, Mendola N, Aquilante CL. Pharmacogenetic testing among patients with depression in a US managed care population. Clin Transl Sci 2022; 15:1644-1653. [PMID: 35385214 PMCID: PMC9283740 DOI: 10.1111/cts.13279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/14/2022] [Accepted: 03/10/2022] [Indexed: 12/01/2022] Open
Abstract
Actionable drug–gene pairs relevant to depression treatment include CYP2D6 and CYP2C19 with specific antidepressants. While clinical use of pharmacogenetic testing is growing, little is known about pharmacogenetic testing for depression treatment in managed care. We determined the incidence of single‐gene CYP2D6 and CYP2C19 testing following a new depression episode among US managed care patients, and described characteristics and antidepressant use of patients receiving tests. We used paid medical and pharmacy claims for patients from commercial health plans in the US. For adult patients with a new depression episode from January 1, 2013 to June 30, 2018, we identified covered claims for single‐gene CYP2D6 and CYP2C19 pharmacogenetic tests and antidepressant fills. Fewer than 1% (n = 1795) of the depressed cohort (n = 438,534) received a single‐gene CYP2D6 or CYP2C19 test through their insurance within 365 days of their earliest depression episode. The percentage of patients who received a test nearly tripled from 0.2% in 2013 to 0.5% in 2014 before plateauing at 0.4% from 2014 to 2017. Among the patients who received a single‐gene CYP2D6 or CYP2C19 test and filled an antidepressant within 365 days of their depression diagnosis, up to 30% may have had their initial antidepressant informed by the test result. Our findings describe the use of antidepressants before and after pharmacogenetic testing, which is clinically relevant as pharmacogenomic testing becomes more common in clinical practice. Our study also emphasizes the need for procedure and billing codes that capture multiple‐gene panel tests to be more widely implemented in administrative databases.
Collapse
Affiliation(s)
- Heather D Anderson
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Thida M Thant
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - David P Kao
- Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kristy R Crooks
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nicholas Mendola
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Christina L Aquilante
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| |
Collapse
|
12
|
The Value of Pharmacogenetics to Reduce Drug-Related Toxicity in Cancer Patients. Mol Diagn Ther 2022; 26:137-151. [PMID: 35113367 PMCID: PMC8975257 DOI: 10.1007/s40291-021-00575-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
Many anticancer drugs cause adverse drug reactions (ADRs) that negatively impact safety and reduce quality of life. The typical narrow therapeutic range and exposure-response relationships described for anticancer drugs make precision dosing critical to ensure safe and effective drug exposure. Germline mutations in pharmacogenes contribute to inter-patient variability in pharmacokinetics and pharmacodynamics of anticancer drugs. Patients carrying reduced-activity or loss-of-function alleles are at increased risk for ADRs. Pretreatment genotyping offers a proactive approach to identify these high-risk patients, administer an individualized dose, and minimize the risk of ADRs. In the field of oncology, the most well-studied gene-drug pairs for which pharmacogenetic dosing recommendations have been published to improve safety are DPYD-fluoropyrimidines, TPMT/NUDT15-thiopurines, and UGT1A1-irinotecan. Despite the presence of these guidelines, the scientific evidence showing the benefits of pharmacogenetic testing (e.g., improved safety and cost-effectiveness) and the development of efficient multi-gene genotyping panels, routine pretreatment testing for these gene-drug pairs has not been implemented widely in the clinic. Important considerations required for widespread clinical implementation include pharmacogenetic education of physicians, availability or allocation of institutional resources to build an efficient clinical infrastructure, international standardization of guidelines, uniform adoption of guidelines by regulatory agencies leading to genotyping requirements in drug labels, and development of cohesive reimbursement policies for pretreatment genotyping. Without clinical implementation, the potential of pharmacogenetics to improve patient safety remains unfulfilled.
Collapse
|
13
|
Liu WY, Chien CW, Tung TH. Healthcare practice strategies for integrating personalized medicine: Management of COVID-19. World J Clin Cases 2021; 9:8647-8657. [PMID: 34734043 PMCID: PMC8546804 DOI: 10.12998/wjcc.v9.i29.8647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/22/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Personalized medicine is the tailor-made clinical treatment to the individual characteristics of each patient. It may be considered an extension of traditional approaches to knowing and treating diseases. Personalized medicine has the potential to change the way of identification and management of health problems. Coronavirus disease 2019 (COVID-19) is an infectious disease that primarily affects the patients' lungs. The first case of pneumonia of unknown cause was reported in Wuhan, China on December 31, 2019. As thus, we are quickly approaching the era of personalized medicine. This review discusses the practices currently used in the management of COVID-19 and how they relate to personalized medicine.
Collapse
Affiliation(s)
- Wen-Yi Liu
- Department of Health Policy and Management, Bloomberg School of Public Health, Johns Hopkins University, MD 21205, United States; Shanghai Bluecross Medical Science Institute, Shanghai 201100, Shanghai Province, China
- Institute for Hospital Management, Tsing Hua University, Shenzhen Campus, Shenzhen 518055, Guangdong Province, China
| | - Ching-Wen Chien
- Institute for Hospital Management, Tsing Hua University, Shenzhen Campus, Shenzhen 518055, Guangdong Province, China
| | - Tao-Hsin Tung
- Evidence-based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| |
Collapse
|
14
|
Margaretos NM, Panzer AD, Lai RC, Sanon M, Michalopoulos E, Redmond AM, Moghadam R, Chambers JD. Variation in health plan coverage of ESAs for anemia due to chronic kidney disease. J Manag Care Spec Pharm 2021; 27:1221-1229. [PMID: 34464213 PMCID: PMC10391084 DOI: 10.18553/jmcp.2021.27.9.1221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND: Because health plans each issue their own policies, drug coverage can vary. This variation can result in patients having unequal access to treatment. In this study, we evaluate commercial health plans' coverage policies for erythropoiesis-stimulating agents (ESAs) for patients with anemia resulting from chronic kidney disease (CKD). OBJECTIVES: To assess how a set of US commercial health plans cover ESAs for patients with anemia due to CKD. Our second objective was to examine the evidence that the plans reviewed when formulating their coverage policies. METHODS: We used the Tufts Medical Center Specialty Drug and Evidence and Coverage Database to identify coverage policies issued by 17 of the largest US commercial health plans for ESAs. The following drugs were indicated for anemia due to CKD: darbepoetin alfa, methoxy polyethylene glycol-epoetin beta, epoetin alfa (available as two brands), and epoetin alfa-epbx. Coverage policies were current as of May 2019. We determined whether the health plans applied any restrictions, such as step therapy protocols or patient subgroup restrictions, in their coverage policies. We categorized the evidence that plans cited to support their policies into seven categories: randomized controlled trials (RCTs), real-world evidence (RWE) studies (studies based on data collected in a real-world setting), other clinical studies (eg, single arm trials), systematic reviews and/or meta-analyses, clinical or treatment guidelines, health technology assessments, and economic evaluations. RESULTS: We categorized 72.5% of coverage policies (58/80 policies) as equivalent to the FDA label and 27.5% (22/80 policies) as more restrictive. In restricted policies, plans most often applied step therapy protocols (18/22 policies), followed by prescriber requirements (4/22 policies), and patient subgroup restrictions (3/22 policies). Five health plans applied restrictions in at least half of their coverage policies; seven plans did not apply restrictions in any policy. Plans that cited evidence reviewed an average of 10 citations across their ESA coverage policies, ranging from one to 29 studies. Plans varied with respect to the types of cited studies: at least 50% of evidence cited by five health plans was RCTs, while half or more of the evidence cited by four health plans was clinical or treatment guidelines. CONCLUSIONS: Health plans varied in how they covered ESAs for patients with anemia due to CKD and in the evidence cited in their coverage policies. Inconsistencies in plans' coverage policies may have implications for patients' access to ESAs. DISCLOSURES: This study was funded by Otsuka Pharmaceutical Development and Commercialization. Sanon, Redmond, and Mogahadam are employed by Otsuka Pharmaceutical. Michalopoulos was employed by Otsuka Pharmaceutical at the time of this study. Margaretos, Panzer, and Chambers are employed by Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Center for the Evaluation of Value and Risk in Health. Lai was with Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Center for the Evaluation of Value and Risk in Health at the time of this study.
Collapse
Affiliation(s)
- NikoLetta M Margaretos
- Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Center for the Evaluation of Value and Risk in Health, Boston, MA
| | - Ari D Panzer
- Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Center for the Evaluation of Value and Risk in Health, Boston, MA
| | - Rachel C Lai
- Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Center for the Evaluation of Value and Risk in Health, Boston, MA
| | - Myrlene Sanon
- Otsuka Pharmaceutical Development & Commercialization, Princeton, NJ
| | | | - Ann-Marie Redmond
- Otsuka Pharmaceutical Development & Commercialization, Princeton, NJ
| | - Reza Moghadam
- Otsuka Pharmaceutical Development & Commercialization, Princeton, NJ
| | - James D Chambers
- Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Center for the Evaluation of Value and Risk in Health, Boston, MA
| |
Collapse
|
15
|
Modell SM, Allen CG, Ponte A, Marcus G. Cancer genetic testing in marginalized groups during an era of evolving healthcare reform. J Cancer Policy 2021; 28:100275. [PMID: 35559905 PMCID: PMC8224823 DOI: 10.1016/j.jcpo.2021.100275] [Citation(s) in RCA: 14] [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: 10/13/2020] [Accepted: 01/31/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND The Affordable Care Act and subsequent reforms pose tradeoffs for racial-ethnic, rural, and sex-related groups in the United States experiencing disparities in BRCA1/2 genetic counseling and testing and colorectal cancer screening, calling for policy changes. METHODS A working group of the American Public Health Association Genomics Forum Policy Committee engaged in monthly meetings to examine ongoing literature and identify policy alternatives in the coverage of cancer genetic services for marginalized groups. 589 items were collected; 408 examined. Efforts continued from February 2015 through September 2020. RESULTS African Americans and Latinos have shown 7-8 % drops in uninsured rates since the Exchanges opened. The ACA has increased BRCA1/2 test availability while several disparities remain, including by sex. Rural testing and screening utilization rates have improved. Medicaid expansion and the inclusion of Medicare in the ACA have resulted in mixed improvements in colorectal cancer screening rates in marginalized groups. CONCLUSION Cancer genetic testing and screening to date have only partially benefited from healthcare reforms. Sensitivity to cost concerns and further monitoring of emerging data are needed. A reduction in disparities depends on the availability of private insurance, Medicaid and Medicare to the marginalized. Attention to value-based design and the way cancer benefits are translated into actual testing and screening are crucial. POLICY SUMMARY The findings suggest the need for further benefits-related health agency interpretation of and amendments to the ACA, continued Medicaid and innovative Medicare expansion, and incorporation of cancer services values-based considerations at several levels, aimed at reducing group disparities.
Collapse
Affiliation(s)
- Stephen M Modell
- Epidemiology, University of Michigan School of Public Health, M5409 SPH II, 1415 Washington Hts., Ann Arbor, MI, 48109, United States.
| | - Caitlin G Allen
- Behavioral, Social and Health Education Sciences, Rollins School of Public Health, Emory University, 1518 Clifton Rd., Atlanta, GA, 30322, United States
| | - Amy Ponte
- Genedu Health Solutions, 47 Petigru Dr., Beaufort, SC, 29902, United States
| | - Gail Marcus
- Genetics and Newborn Screening Unit, North Carolina Department of Health and Human Services, C/O CDSA of the Cape Fear, 3311 Burnt Mill Dr., Wilmington, NC, 28403, United States
| |
Collapse
|
16
|
Implementation of Pharmacogenomics and Artificial Intelligence Tools for Chronic Disease Management in Primary Care Setting. J Pers Med 2021; 11:jpm11060443. [PMID: 34063850 PMCID: PMC8224063 DOI: 10.3390/jpm11060443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic disease management often requires use of multiple drug regimens that lead to polypharmacy challenges and suboptimal utilization of healthcare services. While the rising costs and healthcare utilization associated with polypharmacy and drug interactions have been well documented, effective tools to address these challenges remain elusive. Emerging evidence that proactive medication management, combined with pharmacogenomic testing, can lead to improved health outcomes and reduced cost burdens may help to address such gaps. In this report, we describe informatic and bioanalytic methodologies that integrate weak signals in symptoms and chief complaints with pharmacogenomic analysis of ~90 single nucleotide polymorphic variants, CYP2D6 copy number, and clinical pharmacokinetic profiles to monitor drug–gene pairs and drug–drug interactions for medications with significant pharmacogenomic profiles. The utility of the approach was validated in a virtual patient case showing detection of significant drug–gene and drug–drug interactions of clinical significance. This effort is being used to establish proof-of-concept for the creation of a regional database to track clinical outcomes in patients enrolled in a bioanalytically-informed medication management program. Our integrated informatic and bioanalytic platform can provide facile clinical decision support to inform and augment medication management in the primary care setting.
Collapse
|
17
|
Caffrey AR, Borrelli EP. The art and science of drug titration. Ther Adv Drug Saf 2021; 11:2042098620958910. [PMID: 33796256 PMCID: PMC7967860 DOI: 10.1177/2042098620958910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
A “one-size-fits-all” approach has been the standard for drug dosing, in
particular for agents with a wide therapeutic index. The scientific
principles of drug titration, most commonly used for medications with
a narrow therapeutic index, are to give the patient adequate and
effective treatment, at the lowest dose possible, with the aim of
minimizing unnecessary medication use and side effects. The art of
drug titration involves the interplay of scientific drug titration
principles with the clinical expertise of the healthcare provider, and
an individualized, patient-centered partnership between the provider
and the patient to review the delicate balance of perceived benefits
and risks from both perspectives. Drug titration may occur as up-,
down-, or cross-titration depending on whether the goal is to reach or
maintain a therapeutic outcome, decrease the risk of adverse effects,
or prevent withdrawal/discontinuation syndromes or recurrence of
disease. Drug titration introduces additional complexities surrounding
the conduct of clinical trials and real-world studies, confounding our
understanding of the true effect of medications. In clinical practice,
wide variations in titration schedules may exist due to a lack of
evidence and consensus on titration approaches that achieve an optimal
benefit-harm profile. Further, drug titration may be challenging for
patients to follow, resulting in suboptimal adherence and may require
increased healthcare-related visits and coordination of care amongst
providers. Despite the challenges associated with drug titration, it
is a personalized approach to drug dosing that blends science with
art, and with supportive real-world outcomes-based evidence, can be
effective for optimizing pharmacotherapeutic outcomes and improving
drug safety.
Collapse
Affiliation(s)
- Aisling R Caffrey
- University of Rhode Island College of Pharmacy, 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Eric P Borrelli
- University of Rhode Island College of Pharmacy, Kingston, RI, USA
| |
Collapse
|
18
|
Ayati N, Afzali M, Hasanzad M, Kebriaeezadeh A, Rajabzadeh A, Nikfar S. Pharmacogenomics Implementation and Hurdles to Overcome; In the Context of a Developing Country. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:92-106. [PMID: 35194431 PMCID: PMC8842599 DOI: 10.22037/ijpr.2021.114899.15091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Having multiple dimensions, uncertainties and several stakeholders, the costly pharmacogenomics (PGx) is associated with dynamic implementation complexities. Identification of these challenges is critical to harness its full potential, especially in developing countries with fragile healthcare systems and scarce resources. This is the first study aimed to identify most salient challenges related to PGx implementation, with respect to the experiences of early-adopters and local experts' prospects, in the context of a developing country in the Middle East. To perform a comprehensive reconnaissance on PGx adoption challenges a scoping literature review was conducted based on national drug policy components: efficacy/safety, access, affordability and rational use of medicine (RUM). Strategic option development and analysis workshop method with cognitive mapping as the technique was used to evaluate challenges in the context of Iran. The cognitive maps were face-validated and analyzed via Decision Explorer XML. The findings indicated a complex network of issues relative to PGx adoption, categorized in national drug policy indicators. In the rational use of medicine category, ethics, education, bench -to- bedside strategies, guidelines, compliance, and health system issues were found. Clinical trial issues, test's utility, and biomarker validation were identified in the efficacy group. Affordability included pricing, reimbursement, and value assessment issues. Finally, access category included regulation, availability, and stakeholder management challenges. The current study identified the most significant challenges ahead of clinical implementation of PGx in a developing country. This could be the basis of a policy-note development in future work, which may consolidate vital communication among stakeholders and accelerate the efficient implementation in developing new-comer countries.
Collapse
Affiliation(s)
- Nayyereh Ayati
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Monireh Afzali
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Mandana Hasanzad
- Medical Genomics Research Center, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Abbas Kebriaeezadeh
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Ali Rajabzadeh
- Department of Department of Industrial Management, Faculty of Management and Economics, Tarbiat Modares University, Tehran, Iran.
| | - Shekoufeh Nikfar
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Ward ET, Kostick KM, Lázaro-Muñoz G. Integrating Genomics into Psychiatric Practice: Ethical and Legal Challenges for Clinicians. Harv Rev Psychiatry 2020; 27:53-64. [PMID: 30614887 PMCID: PMC6326091 DOI: 10.1097/hrp.0000000000000203] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Psychiatric genomics is a rapidly growing field that holds much promise for improving risk prediction, prevention, diagnosis, treatment selection, and understanding of the pathogenesis of patients' symptoms. The field of psychiatry (i.e., professional organizations, mental health clinicians, educational institutions), however, needs to address numerous challenges to promote the responsible translation of genomic technologies and knowledge into psychiatric practice. The goal of this article is to review how clinicians currently encounter and use genomics in the clinic, to summarize the existing literature on how clinicians feel about the use of genomics in psychiatry, and to analyze foreseeable ethical and legal challenges for the responsible integration of genomics into psychiatric care at the structural and clinic levels. Structural challenges are defined as aspects of the larger system of psychiatric practice that constitute potential barriers to the responsible integration of genomics for the purposes of psychiatric care and prevention. These structural challenges exist at a level where professional groups can intervene to set standards and regulate the practice of psychiatry and genomics. Clinic-level challenges are day-to-day issues clinicians face when managing genomic tests in the clinic. We discuss the need for action to mitigate these challenges and maximize the clinical and social utility of psychiatric genomics, including the following: expanding genomics training among mental health clinicians; establishing practice guidelines that consider potential clinical, psychological, and social implications of psychiatric genomics; promoting an integrated care model for managing genomics in psychiatry; emphasizing patient engagement and informed consent when managing genomic testing in psychiatric care.
Collapse
Affiliation(s)
- Eric T Ward
- From the University of North Carolina School of Medicine (Dr. Ward); Center for Medical Ethics and Health Policy, Baylor College of Medicine (Drs. Kostick and Lázaro-Muñoz)
| | | | | |
Collapse
|
20
|
Huebner T, Steffens M, Linder R, Fracowiak J, Langner D, Garling M, Falkenberg F, Roethlein C, Gomm W, Haenisch B, Stingl J. Influence of metabolic profiles on the safety of drug therapy in routine care in Germany: protocol of the cohort study EMPAR. BMJ Open 2020; 10:e032624. [PMID: 32345696 PMCID: PMC7213853 DOI: 10.1136/bmjopen-2019-032624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Pre-emptive testing of pharmacogenetically relevant single-nucleotide polymorphisms can be an effective tool in the prevention of adverse drug reactions and therapy resistance. However, most of the tests are not used as standard in routine care in Germany because of lacking evidence for the clinical and economical benefit and their impact on the usage of healthcare services. We address this issue by investigating the influence of pharmacogenetic profiles on the use of healthcare services over an extended period of several years using routine care data from a statutory health insurance company. The goal is to provide clinical evidence whether pre-emptive pharmacogenetic testing of metabolic profiles in routine care in Germany is beneficial and cost-effective. METHODS AND ANALYSIS The EMPAR (Einfluss metabolischer Profile auf die Arzneimitteltherapiesicherheit in der Routineversorgung) study is a non-interventional cohort study conducted to analyse pharmacogenetic risk factors that are important for drug therapy by means of endpoints relevant for healthcare. The analysis is based on pharmacogenetic profiles and statutory health insurance data. We perform pharmacogenetic, pharmacoepidemiological and pharmacoeconomic analyses using health care utilisation scores and machine learning techniques. Therefore, we aim to include about 10 000 patients (≥18 years) insured by the health insurance provider Techniker Krankenkasse. The study focuses on patients with prescriptions of anticoagulants and prescriptions of cholesterol-lowering drugs. Also, a screening for special pharmacogenetic characteristics will be performed in patients with at least one Y57.9! diagnosis (Complication of medical and surgical care: drug or medicament, unspecified). Outcomes include the utilisation of health insurance services, the incidence of incapacity for work and costs for drugs and treatment. ETHICS AND DISSEMINATION The protocol was approved by the Ethics Committee of the Medical Faculty, University of Bonn (Lfd. Nr. 339/17). The results of this research project will be published in scientific open access journals and at conferences. TRIAL REGISTRATION NUMBER German Clinical Trials Register, DRKS00013909.
Collapse
Affiliation(s)
- Tatjana Huebner
- Research Division, Federal Institute for Drugs and Medical Devices, Bonn, North Rhine-Westphalia, Germany
| | - Michael Steffens
- Research Division, Federal Institute for Drugs and Medical Devices, Bonn, North Rhine-Westphalia, Germany
| | | | - Jochen Fracowiak
- Research Division, Federal Institute for Drugs and Medical Devices, Bonn, North Rhine-Westphalia, Germany
| | | | | | | | - Christoph Roethlein
- Population Health Sciences, German Centre for Neurodegenerative Diseases, Bonn, North Rhine-Westphalia, Germany
| | - Willy Gomm
- Population Health Sciences, German Centre for Neurodegenerative Diseases, Bonn, North Rhine-Westphalia, Germany
| | - Britta Haenisch
- Research Division, Federal Institute for Drugs and Medical Devices, Bonn, North Rhine-Westphalia, Germany
- Population Health Sciences, German Centre for Neurodegenerative Diseases, Bonn, North Rhine-Westphalia, Germany
- Centre for Translational Medicine, University of Bonn, Bonn, North Rhine-Westphalia, Germany
| | - Julia Stingl
- Institute for Clinical Pharmacology, RWTH Aachen University, Aachen, North Rhine-Westphalia, Germany
| |
Collapse
|
21
|
Wu AC, Kiley JP, Noel PJ, Amur S, Burchard EG, Clancy JP, Galanter J, Inada M, Jones TK, Kropski JA, Loyd JE, Nogee LM, Raby BA, Rogers AJ, Schwartz DA, Sin DD, Spira A, Weiss ST, Young LR, Himes BE. Current Status and Future Opportunities in Lung Precision Medicine Research with a Focus on Biomarkers. An American Thoracic Society/National Heart, Lung, and Blood Institute Research Statement. Am J Respir Crit Care Med 2019; 198:e116-e136. [PMID: 30640517 DOI: 10.1164/rccm.201810-1895st] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Thousands of biomarker tests are either available or under development for lung diseases. In many cases, adoption of these tests into clinical practice is outpacing the generation and evaluation of sufficient data to determine clinical utility and ability to improve health outcomes. There is a need for a systematically organized report that provides guidance on how to understand and evaluate use of biomarker tests for lung diseases. METHODS We assembled a diverse group of clinicians and researchers from the American Thoracic Society and leaders from the National Heart, Lung, and Blood Institute with expertise in various aspects of precision medicine to review the current status of biomarker tests in lung diseases. Experts summarized existing biomarker tests that are available for lung cancer, pulmonary arterial hypertension, idiopathic pulmonary fibrosis, asthma, chronic obstructive pulmonary disease, sepsis, acute respiratory distress syndrome, cystic fibrosis, and other rare lung diseases. The group identified knowledge gaps that future research studies can address to efficiently translate biomarker tests into clinical practice, assess their cost-effectiveness, and ensure they apply to diverse, real-life populations. RESULTS We found that the status of biomarker tests in lung diseases is highly variable depending on the disease. Nevertheless, biomarker tests in lung diseases show great promise in improving clinical care. To efficiently translate biomarkers into tests used widely in clinical practice, researchers need to address specific clinical unmet needs, secure support for biomarker discovery efforts, conduct analytical and clinical validation studies, ensure tests have clinical utility, and facilitate appropriate adoption into routine clinical practice. CONCLUSIONS Although progress has been made toward implementation of precision medicine for lung diseases in clinical practice in certain settings, additional studies focused on addressing specific unmet clinical needs are required to evaluate the clinical utility of biomarkers; ensure their generalizability to diverse, real-life populations; and determine their cost-effectiveness.
Collapse
|
22
|
Lee J, Cheng N, Tai H, Jimmy Juang J, Wu C, Lin L, Hwang J, Lin J, Chiang F, Tsai C. CYP2C19 Polymorphism is Associated With Amputation Rates in Patients Taking Clopidogrel After Endovascular Intervention for Critical Limb Ischaemia. Eur J Vasc Endovasc Surg 2019; 58:373-382. [DOI: 10.1016/j.ejvs.2019.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 02/12/2019] [Indexed: 02/08/2023]
|
23
|
Bush WS, Cooke Bailey JN, Beno MF, Crawford DC. Bridging the Gaps in Personalized Medicine Value Assessment: A Review of the Need for Outcome Metrics across Stakeholders and Scientific Disciplines. Public Health Genomics 2019; 22:16-24. [PMID: 31454805 PMCID: PMC6752968 DOI: 10.1159/000501974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/07/2019] [Indexed: 12/14/2022] Open
Abstract
Despite monumental advances in genomics, relatively few health care provider organizations in the United States offer personalized or precision medicine as part of the routine clinical workflow. The gaps between research and applied genomic medicine may be a result of a cultural gap across various stakeholders representing scientists, clinicians, patients, policy makers, and third party payers. Scientists are trained to assess the health care value of genomics by either quantifying population-scale effects, or through the narrow lens of clinical trials where the standard of care is compared with the predictive power of a single or handful of genetic variants. While these metrics are an essential first step in assessing and documenting the clinical utility of genomics, they are rarely followed up with other assessments of health care value that are critical to stakeholders who use different measures to define value. The limited value assessment in both the research and implementation science of precision medicine is likely due to necessary logistical constraints of these teams; engaging bioethicists, health care economists, and individual patient belief systems is incredibly daunting for geneticists and informaticians conducting research. In this narrative review, we concisely describe several definitions of value through various stakeholder viewpoints. We highlight the existing gaps that prevent clinical translation of scientific findings generally as well as more specifically using two present-day, extreme scenarios: (1) genetically guided warfarin dosing representing a handful of genetic markers and more than 10 years of basic and translational research, and (2) next-generation sequencing representing genome-dense data lacking substantial evidence for implementation. These contemporary scenarios highlight the need for various stakeholders to broadly adopt frameworks designed to define and collect multiple value measures across different disciplines to ultimately impact more universal acceptance of and reimbursement for genomic medicine.
Collapse
Affiliation(s)
- William S Bush
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jessica N Cooke Bailey
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark F Beno
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana C Crawford
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA,
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA,
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA,
| |
Collapse
|
24
|
Sivadas A, Scaria V. Population-scale genomics-Enabling precision public health. ADVANCES IN GENETICS 2018; 103:119-161. [PMID: 30904093 DOI: 10.1016/bs.adgen.2018.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The current excitement for affordable genomics technologies and national precision medicine initiatives marks a turning point in worldwide healthcare practices. The last decade of global population sequencing efforts has defined the enormous extent of genetic variation in the human population resulting in insights into differential disease burden and response to therapy within and between populations. Population-scale pharmacogenomics helps to provide insights into the choice of optimal therapies and an opportunity to estimate, predict and minimize adverse events. Such an approach can potentially empower countries to formulate national selection and dosing policies for therapeutic agents thereby promoting public health with precision. We review the breadth and depth of worldwide population-scale sequencing efforts and its implications for the implementation of clinical pharmacogenetics toward making precision medicine a reality.
Collapse
Affiliation(s)
- Ambily Sivadas
- GN Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Vinod Scaria
- GN Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
25
|
Gap between the US and Japan in coverage of pharmacogenomic biomarkers by health insurance programs: More coverage is needed in Japan. Drug Metab Pharmacokinet 2018; 33:243-249. [DOI: 10.1016/j.dmpk.2018.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023]
|
26
|
EVIDENCE REQUIRED BY HEALTH TECHNOLGY ASSESSMENT AND REIMBURSEMENT BODIES EVALUATING DIAGNOSTIC OR PROGNOSTIC ALGORITHMS THAT INCLUDE OMICS DATA. Int J Technol Assess Health Care 2018; 34:368-377. [PMID: 30136642 DOI: 10.1017/s026646231800048x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Multi-analyte assays with algorithmic analyses (MAAAs) use combinations of circulating and clinical markers including omics-based sources for diagnostic and/or prognostic purposes. Assessing MAAAs is challenging under existing health technology assessment (HTA) methods or practices. We undertook a scoping review to explore the HTA methods used for MAAAs to identify the criteria used for clinical research and reimbursement purposes. METHODS This review included only non-companion (stand-alone) tests that are actionable and that have been evaluated by leading HTA or insurer/reimbursement bodies up to September 2017. RESULTS Twenty-five reports and articles evaluating seventeen MAAAs were examined, most of which have been developed in oncology. The two main models used were the EUnetHTA Core model and the Evaluation of Genomic Applications in Practice and Prevention ACCE framework. Clinical validity and utility criteria were used, as were economic, ethical, legal, and social aspects. Economic evidence on MAAAs was scarce, and there is no consensus on whether the perspectives used are sufficiently broad to include all relevant stakeholders. CONCLUSIONS Clinical utility and efficiency were the most used criteria, with stronger evidence needed linking the use of the algorithm with the clinical outcomes in real-life practice. HTA bodies must as well consider questions related to the analytical validity of MAAAs or with organizational aspects. The two main models, the EUnetHTA Core model and the ACCE framework, could be adapted to the assessment of MAAAs.
Collapse
|
27
|
Insurance Coverage Policies for Pharmacogenomic and Multi-Gene Testing for Cancer. J Pers Med 2018; 8:jpm8020019. [PMID: 29772692 PMCID: PMC6023380 DOI: 10.3390/jpm8020019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/17/2022] Open
Abstract
Insurance coverage policies are a major determinant of patient access to genomic tests. The objective of this study was to examine differences in coverage policies for guideline-recommended pharmacogenomic tests that inform cancer treatment. We analyzed coverage policies from eight Medicare contractors and 10 private payers for 23 biomarkers (e.g., HER2 and EGFR) and multi-gene tests. We extracted policy coverage and criteria, prior authorization requirements, and an evidence basis for coverage. We reviewed professional society guidelines and their recommendations for use of pharmacogenomic tests. Coverage for KRAS, EGFR, and BRAF tests were common across Medicare contractors and private payers, but few policies covered PML/RARA, CD25, or G6PD. Twelve payers cover at least one multi-gene test for nonsmall cell lung cancer, citing emerging clinical recommendations. Coverage policies for single and multi-gene tests for cancer treatments are relatively consistent among Medicare contractors despite the lack of national coverage determinations. In contrast, coverage for these tests varied across private payers. Patient access to tests is governed by prior authorization among eight private payers. Substantial variations in how payers address guideline-recommended pharmacogenomic tests and the common use of prior authorization underscore the need for additional studies of the effects of coverage variation on cancer care and patient outcomes.
Collapse
|
28
|
Rodieux F, Vutskits L, Posfay-Barbe KM, Habre W, Piguet V, Desmeules JA, Samer CF. When the Safe Alternative Is Not That Safe: Tramadol Prescribing in Children. Front Pharmacol 2018; 9:148. [PMID: 29556194 PMCID: PMC5844975 DOI: 10.3389/fphar.2018.00148] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/13/2018] [Indexed: 01/10/2023] Open
Abstract
Children represent a vulnerable population in which management of nociceptive pain is complex. Drug responses in children differ from adults due to age-related differences. Moreover, therapeutic choices are limited by the lack of indication for a number of analgesic drugs due to the challenge of conducting clinical trials in children. Furthermore the assessment of efficacy as well as tolerance may be complicated by children's inability to communicate properly. According to the World Health Organization, weak opioids such as tramadol and codeine, may be used in addition to paracetamol and ibuprofen for moderate nociceptive pain in both children and adults. However, codeine prescription has been restricted for the last 5 years in children because of the risk of fatal overdoses linked to the variable activity of cytochrome P450 (CYP) 2D6 which bioactivates codeine. Even though tramadol has been considered a safe alternative to codeine, it is well established that tramadol pharmacodynamic opioid effects, efficacy and safety, are also largely influenced by CYP2D6 activity. For this reason, the US Food and Drug Administration recently released a boxed warning regarding the use of tramadol in children. To provide safe and effective tramadol prescription in children, a personalized approach, with dose adaptation according to CYP2D6 activity, would certainly be the safest method. We therefore recommend this approach in children requiring chronic or recurrent nociceptive pain treatment with tramadol. In case of acute inpatients nociceptive pain management, prescribing tramadol at the minimal effective dose, in a child appropriate dosage form and after clear instructions are given to the parents, remains reasonable based on current data. In all other situations, morphine should be preferred for moderate to severe nociceptive pain conditions.
Collapse
Affiliation(s)
- Frédérique Rodieux
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Laszlo Vutskits
- Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland.,Department of Basic Neuroscience, Faculty of Medicine, University of GenevaGeneva, Switzerland.,Division of Anesthesiology, Unit for Pediatric Anesthesia, Children's Hospitals of Geneva, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Klara M Posfay-Barbe
- Pediatric Infectious Diseases Unit, Department of Pediatrics, Children's Hospital of Geneva, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Walid Habre
- Division of Anesthesiology, Unit for Pediatric Anesthesia, Children's Hospitals of Geneva, Geneva University Hospitals, University of GenevaGeneva, Switzerland.,Anesthesiological Investigations Unit, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Valérie Piguet
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Jules A Desmeules
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of LausanneGeneva, Switzerland
| | - Caroline F Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| |
Collapse
|
29
|
Keeling NJ, Rosenthal MM, West-Strum D, Patel AS, Haidar CE, Hoffman JM. Preemptive pharmacogenetic testing: exploring the knowledge and perspectives of US payers. Genet Med 2017; 21:1224-1232. [PMID: 31048813 PMCID: PMC5920773 DOI: 10.1038/gim.2017.181] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/14/2017] [Indexed: 01/06/2023] Open
Abstract
PURPOSE Preemptive pharmacogenetic testing aims to optimize medication use by having genetic information at the point of prescribing. Payers’ decisions influence implementation of this technology. We investigated U.S. payers’ knowledge, awareness, and perspectives on preemptive pharmacogenetic testing. METHODS A qualitative study was conducted using semi-structured interviews. Participants were screened for eligibility through an online survey. A blended inductive and deductive approach was used to analyze the transcripts. Two authors conducted an iterative reading process to code and categorize the data. RESULTS Medical or pharmacy directors from 14 payer organizations covering 122 million U.S. lives were interviewed. Three concept domains and ten dimensions were developed. Key findings include: clinical utility concerns and limited exposure to preemptive germline testing, continued preference for outcomes from randomized controlled trials, interest in guideline development, importance of demonstrating an impact on clinical decision making, concerns of downstream costs and benefit predictability, and the impact of public stakeholders such as the FDA and CMS. CONCLUSION Both barriers and potential facilitators exist to developing cohesive reimbursement policy for pharmacogenetics, and there are unique challenges for the preemptive testing model. Prospective outcome studies, more precisely defining target populations, and predictive economic models are important considerations for future research.
Collapse
Affiliation(s)
- Nicholas J Keeling
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, Oxford, Mississippi, USA.,Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Meagen M Rosenthal
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, Oxford, Mississippi, USA
| | - Donna West-Strum
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, Oxford, Mississippi, USA
| | - Amit S Patel
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, Oxford, Mississippi, USA.,Medical Marketing Economics, Oxford, Mississippi, USA
| | - Cyrine E Haidar
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - James M Hoffman
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
30
|
EXAMINING EVIDENCE IN U.S. PAYER COVERAGE POLICIES FOR MULTI-GENE PANELS AND SEQUENCING TESTS. Int J Technol Assess Health Care 2017; 33:534-540. [PMID: 29065945 DOI: 10.1017/s0266462317000903] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVES The aim of this study was to examine the evidence payers cited in their coverage policies for multi-gene panels and sequencing tests (panels), and to compare these findings with the evidence payers cited in their coverage policies for other types of medical interventions. METHODS We used the University of California at San Francisco TRANSPERS Payer Coverage Registry to identify coverage policies for panels issued by five of the largest US private payers. We reviewed each policy and categorized the evidence cited within as: clinical studies, systematic reviews, technology assessments, cost-effectiveness analyses (CEAs), budget impact studies, and clinical guidelines. We compared the evidence cited in these coverage policies for panels with the evidence cited in policies for other intervention types (pharmaceuticals, medical devices, diagnostic tests and imaging, and surgical interventions) as reported in a previous study. RESULTS Fifty-five coverage policies for panels were included. On average, payers cited clinical guidelines in 84 percent of their coverage policies (range, 73-100 percent), clinical studies in 69 percent (50-87 percent), technology assessments 47 percent (33-86 percent), systematic reviews or meta-analyses 31 percent (7-71 percent), and CEAs 5 percent (0-7 percent). No payers cited budget impact studies in their policies. Payers less often cited clinical studies, systematic reviews, technology assessments, and CEAs in their coverage policies for panels than in their policies for other intervention types. Payers cited clinical guidelines in a comparable proportion of policies for panels and other technology types. CONCLUSIONS Payers in our sample less often cited clinical studies and other evidence types in their coverage policies for panels than they did in their coverage policies for other types of medical interventions.
Collapse
|
31
|
Moyer AM, Caraballo PJ. The challenges of implementing pharmacogenomic testing in the clinic. Expert Rev Pharmacoecon Outcomes Res 2017; 17:567-577. [PMID: 28949250 DOI: 10.1080/14737167.2017.1385395] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Pharmacogenomic testing has the potential to greatly benefit patients by enabling personalization of medication management, ensuring better efficacy and decreasing the risk of side effects. However, to fully realize the potential of pharmacogenomic testing, there are several important issues that must be addressed. Areas covered: In this expert review we discuss current challenges impacting the implementation of pharmacogenomic testing in the clinical practice. We emphasize issues related to testing methods, reporting of the results, test selection, clinical interpretation of the results, cost-effectiveness, and the long-term use of pharmacogenomic results in clinical practice. We identify opportunities and future directions to facilitate clinical implementation. Expert commentary: Several key elements are necessary to optimally integrate pharmacogenomic testing into clinical practice. Collaborative efforts among laboratories are needed to improve standardization of testing and reporting of the results. Clinicians need educational opportunities to improve understanding of which test to order and how to interpret the results. The electronic health records and other clinical systems need to improve their storage of the pharmacogenomics test results and interoperability to facilitate the use of clinically actionable results to improve patient care.
Collapse
Affiliation(s)
- Ann M Moyer
- a Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA
| | - Pedro J Caraballo
- b Department of Medicine , Mayo Clinic , Rochester , MN , USA.,c Center for Translational Informatics and Knowledge Management, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
32
|
Tavakolpour S. Towards personalized medicine for patients with autoimmune diseases: Opportunities and challenges. Immunol Lett 2017; 190:130-138. [DOI: 10.1016/j.imlet.2017.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
|
33
|
A review of international coverage and pricing strategies for personalized medicine and orphan drugs. Health Policy 2017; 121:1240-1248. [PMID: 29033060 DOI: 10.1016/j.healthpol.2017.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 08/12/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Personalized medicine and orphan drugs share many characteristics-both target small patient populations, have uncertainties regarding efficacy and safety at payer submission, and frequently have high prices. Given personalized medicine's rising importance, this review summarizes international coverage and pricing strategies for personalized medicine and orphan drugs as well as their impact on therapy development incentives, payer budgets, and therapy access and utilization. METHODS PubMed, Health Policy Reference Center, EconLit, Google Scholar, and references were searched through February 2017 for articles presenting primary data. RESULTS Sixty-nine articles summarizing 42 countries' strategies were included. Therapy evaluation criteria varied between countries, as did patient cost-share. Payers primarily valued clinical effectiveness; cost was only considered by some. These differences result in inequities in orphan drug access, particularly in smaller and lower-income countries. The uncertain reimbursement process hinders diagnostic testing. Payer surveys identified lack of comparative effectiveness evidence as a chief complaint, while manufacturers sought more clarity on payer evidence requirements. Despite lack of strong evidence, orphan drugs largely receive positive coverage decisions, while personalized medicine diagnostics do not. CONCLUSIONS As more personalized medicine and orphan drugs enter the market, registries can provide better quality evidence on their efficacy and safety. Payers need systematic assessment strategies that are communicated with more transparency. Further studies are necessary to compare the implications of different payer approaches.
Collapse
|
34
|
Vo TT, Bell GC, Owusu Obeng A, Hicks JK, Dunnenberger HM. Pharmacogenomics Implementation: Considerations for Selecting a Reference Laboratory. Pharmacotherapy 2017; 37:1014-1022. [DOI: 10.1002/phar.1985] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Teresa T. Vo
- Department of Pharmacotherapeutics and Clinical Research; College of Pharmacy; University of South Florida; Tampa Florida
| | - Gillian C. Bell
- Personalized Medicine Program; Mission Health; Asheville North Carolina
| | - Aniwaa Owusu Obeng
- The Charles Bronfman Institute for Personalized Medicine; Icahn School of Medicine at Mount Sinai; New York New York
- Pharmacy Department; The Mount Sinai Hospital; New York New York
| | - J. Kevin Hicks
- Division of Population, Science; Department of Individualized Cancer Management; DeBartolo Family Personalized Medicine Institute; Moffitt Cancer Center & Research Institute; Tampa Florida
| | - Henry M. Dunnenberger
- Center for Molecular Medicine; NorthShore University HealthSystem; Evanston Illinois
| |
Collapse
|
35
|
Ciarleglio AE, Ma C. The Daniel K. Inouye College of Pharmacy Scripts: Precision Medicine Through the Use of Pharmacogenomics: Current Status and Barriers to Implementation. HAWAI'I JOURNAL OF MEDICINE & PUBLIC HEALTH : A JOURNAL OF ASIA PACIFIC MEDICINE & PUBLIC HEALTH 2017; 76:265-269. [PMID: 28900583 PMCID: PMC5592383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The precision medicine initiative brought forth by President Barack Obama in 2015 is an important step on the journey to truly personalized medicine. A broad knowledge and understanding of the implications of the pharmacogenomic literature will be critical to the achievement of this goal. While a great amount of data has been published in the areas of pharmacogenomics and pharmacogenetics, there are still relatively few instances in which the need for clinical intervention can be stated without doubt, and which are widely accepted and practiced by the medical community. As our knowledge base rapidly expands, issues such as insurance reimbursement for genetic testing and education of the health care workforce will be paramount to achieving the goal of precision medicine for all patients.
Collapse
Affiliation(s)
- Anita E Ciarleglio
- Assistant Specialist, University of Hawai'i at Hilo, Daniel K. Inouye College of Pharmacy, Hilo, HI
| | - Carolyn Ma
- Dr. Ma is a Board Certified Oncology Pharmacy Specialist with experiences in health systems administration and pharmacy academe
| |
Collapse
|
36
|
Chambers JD, Wilkinson CL, Anderson JE, Chenoweth MD. Variation in Private Payer Coverage of Rheumatoid Arthritis Drugs. J Manag Care Spec Pharm 2017; 22:1176-81. [PMID: 27668566 PMCID: PMC10397684 DOI: 10.18553/jmcp.2016.22.10.1176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Payers in the United States issue coverage determinations to guide how their enrolled beneficiaries use prescription drugs. Because payers create their own coverage policies, how they cover drugs can vary, which in turn can affect access to care by beneficiaries. OBJECTIVE To examine how the largest private payers based on membership cover drugs indicated for rheumatoid arthritis and to determine what evidence the payers reported reviewing when formulating their coverage policies. METHODS Coverage policies issued by the 10 largest private payers that make their policies publicly available were identified for rheumatoid arthritis drugs. Each coverage determination was compared with the drug's corresponding FDA label and categorized according to the following: (a) consistent with the label, (b) more restrictive than the label, (c) less restrictive than the label, or (d) mixed (i.e., more restrictive than the label in one way but less restrictive in another). Each coverage determination was also compared with the American College of Rheumatology (ACR) 2012 treatment recommendations and categorized using the same relative restrictiveness criteria. The policies were then reviewed to identify the evidence that the payers reported reviewing. The identified evidence was divided into the following 6 categories: randomized controlled trials; other clinical studies (e.g., observational studies); health technology assessments; clinical reviews; cost-effectiveness analyses; and clinical guidelines. RESULTS Sixty-nine percent of coverage determinations were more restrictive than the corresponding FDA label; 15% were consistent; 3% were less restrictive; and 13% were mixed. Thirty-four percent of coverage determinations were consistent with the ACR recommendations, 33% were more restrictive; 17% were less restrictive; and 17% were mixed. Payers most often reported reviewing randomized controlled trials for their coverage policies (an average of 2.3 per policy). The payers reported reviewing an average of 1.4 clinical guidelines, 1.1 clinical reviews, 0.8 other clinical studies, and 0.5 technology assessments per policy. Only 1 payer reported reviewing cost-effectiveness analyses. The evidence base that the payers reported reviewing varied in terms of volume and composition. CONCLUSIONS Payers most often covered rheumatoid arthritis drugs more restrictively than the corresponding FDA label indication and the ACR treatment recommendations. Payers reported reviewing a varied evidence base in their coverage policies. DISCLOSURES Funding for this study was provided by Genentech. Chambers has participated in a Sanofi advisory board, unrelated to this study. The authors report no other potential conflicts of interest. Study concept and design were contributed by Chambers. Anderson, Wilkinson, and Chenoweth collected the data, assisted by Chambers, and data interpretation was primarily performed by Chambers, along with Anderson and with assistance from Wilkinson and Chenoweth. The manuscript was written primarily by Chambers, along with Wilkinson and with assistance from Anderson and Chenoweth. Chambers, Chenoweth, Wilkinson, and Anderson revised the manuscript.
Collapse
Affiliation(s)
- James D Chambers
- 1 Tufts University School of Medicine and Center for the Evaluation of Value and Risk in Health, Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts
| | - Colby L Wilkinson
- 2 Center for the Evaluation of Value and Risk in Health, Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts
| | - Jordan E Anderson
- 2 Center for the Evaluation of Value and Risk in Health, Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts
| | - Matthew D Chenoweth
- 2 Center for the Evaluation of Value and Risk in Health, Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
37
|
|
38
|
Mirabbasi SA, Khalighi K, Wu Y, Walker S, Khalighi B, Fan W, Kodali A, Cheng G. CYP2C19 genetic variation and individualized clopidogrel prescription in a cardiology clinic. J Community Hosp Intern Med Perspect 2017; 7:151-156. [PMID: 28808507 PMCID: PMC5538219 DOI: 10.1080/20009666.2017.1347475] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/08/2017] [Indexed: 11/05/2022] Open
Abstract
Background: Clopidogrel (Plavix) is an antiplatelet medication that is routinely used in patients with cardiovascular disease. Cytochrome P2C19 enzymes play a major role in its metabolism, which determines its varied therapeutic level and its effectiveness. Objectives: To customize clopidogrel therapy and evaluate its efficacy by using CYP2C19 genotypic and phenotypic information to improve clinical outcomes in patients. Methods: A total of 465 patients with underlying cardiovascular disease were selected from our out-patient cardiology clinic. DNA sequences of CYP2C19 were analyzed in 465 patients. Results: Of 465 patients, 183 were wild-type homozygous (*1/*1) and 18.8% gain-of function and 19.8% loss-of-function alleles in our patient population The following changes were made: 1) Switching to prasugrel in patients whose genotype noted them to be “Slow metabolizers. This medication adjustment improved clinical outcomes in this patient group. 2) Discontinuing or lowering clopidogrel doses in patients whose genotypes noted them to be “Fast or ultra-fast metabolizes” to decrease bleeding risk. For those who were not on clopidogrel but carried abnormal allele(s), “clopidogrel caution” was documented. These individuals were followed up for 3 years and there has not been any cardiac clinical symptoms, cardiac death or excessive bleeding reported. Conclusions: Given the varied effectiveness of clopidogrel due to its metabolism by CYP2C19 enzyme, and the relatively high frequency of both gain-of-function (18.8%) and loss-of-function (19.8%) alleles in our patient population, we believe that genotyping CYP2C19 is clinically important in order to improve patient outcomes and minimize patient risk.
Collapse
Affiliation(s)
- Seyed Abbas Mirabbasi
- Department of Medicine, Easton Hospital, Drexel University College of Medicine, Easton, PA, USA.,Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| | - Koroush Khalighi
- Department of Medicine, Easton Hospital, Drexel University College of Medicine, Easton, PA, USA.,Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| | - Yin Wu
- Department of Medicine, Easton Hospital, Drexel University College of Medicine, Easton, PA, USA.,Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| | - Stanley Walker
- Department of Medicine, Easton Hospital, Drexel University College of Medicine, Easton, PA, USA
| | - Bahar Khalighi
- School of Pharmacy, Temple University, Philadelphia, PA, USA.,Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| | - Wuqiang Fan
- Department of Medicine, Easton Hospital, Drexel University College of Medicine, Easton, PA, USA.,Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| | - Archana Kodali
- Department of Medicine, Easton Hospital, Drexel University College of Medicine, Easton, PA, USA.,Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| | - Gang Cheng
- Easton Cardiovascular Associates, Cardiovascular Institute, Easton, PA, USA
| |
Collapse
|
39
|
Phillips KA, Deverka PA, Trosman JR, Douglas MP, Chambers JD, Weldon CB, Dervan AP. Payer coverage policies for multigene tests. Nat Biotechnol 2017; 35:614-617. [PMID: 28700544 PMCID: PMC5553867 DOI: 10.1038/nbt.3912] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Kathryn A Phillips
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
- Philip R. Lee Institute for Health Policy, University of California San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Patricia A Deverka
- American Institutes for Research, Chapel Hill, North Carolina, USA
- University of North Carolina, Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, Chapel Hill, North Carolina, USA
| | - Julia R Trosman
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
- Center for Business Models in Healthcare, Chicago, Illinois, USA
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael P Douglas
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
| | - James D Chambers
- The Center for the Evaluation of Value and Risk in Health, Institute for Clinical Research and Health Policy Studies
- Tufts Medical Center, Boston, Massachusetts, USA
| | - Christine B Weldon
- Center for Business Models in Healthcare, Chicago, Illinois, USA
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andrew P Dervan
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
40
|
Messner DA, Koay P, Al Naber J, Cook-Deegan R, Majumder M, Javitt G, Dvoskin R, Bollinger J, Curnutte M, McGuire AL. Barriers to clinical adoption of next-generation sequencing: a policy Delphi panel's solutions. Per Med 2017; 14:339-354. [PMID: 29230253 DOI: 10.2217/pme-2016-0104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aim Identify solutions to the most important policy barriers to the clinical adoption of next-generation sequencing. Materials & methods Four-round modified policy Delphi with a multistakeholder panel of 48 experts. The panel deliberated policy solutions to (previously reported) challenges deemed most important to address. Results The group advocated using consensus panels to promote consistency in payer policies and to standardize test reporting, and favored making genomic data-sharing a condition of regulatory clearance, certification, or accreditation processes. They were split on the role of US FDA. Conclusion Panelists found common ground on solutions for health plan coverage policy consistency, data-sharing, and standardizing reporting, but were sharply divided on the role of the FDA in mitigating risks to patients.
Collapse
Affiliation(s)
- Donna A Messner
- Center for Medical Technology Policy, Baltimore, MD 21202, USA
| | - Pei Koay
- Center for Medical Technology Policy, Baltimore, MD 21202, USA
| | | | - Robert Cook-Deegan
- School for the Future of Innovation in Society, and Consortium for Science, Policy & Outcomes, Arizona State University, Tempe, AZ 85281, USA
| | - Mary Majumder
- Center for Medical Ethics & Health Policy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gail Javitt
- Johns Hopkins Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Rachel Dvoskin
- Johns Hopkins Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Juli Bollinger
- Johns Hopkins Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Margaret Curnutte
- Center for Medical Ethics & Health Policy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amy L McGuire
- Center for Medical Ethics & Health Policy, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
41
|
Dervan AP, Deverka PA, Trosman JR, Weldon CB, Douglas MP, Phillips KA. Payer decision making for next-generation sequencing-based genetic tests: insights from cell-free DNA prenatal screening. Genet Med 2017; 19:559-567. [PMID: 27657682 PMCID: PMC5362360 DOI: 10.1038/gim.2016.145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Cell-free DNA (cfDNA) prenatal screening tests have been rapidly adopted into clinical practice, due in part to positive insurance coverage. We evaluated the framework payers used in making coverage decisions to describe a process that should be informative for other sequencing tests. METHODS We analyzed coverage policies from the 19 largest US private payers with publicly available policies through February 2016, building from the University of California San Francisco TRANSPERS Payer Coverage Policy Registry. RESULTS All payers studied cover cfDNA screening for detection of trisomies 21, 18, and 13 in high-risk, singleton pregnancies, based on robust clinical validity (CV) studies and modeled evidence of clinical utility (CU). Payers typically evaluated the evidence for each chromosomal abnormality separately, although results are offered as part of a panel. Starting in August 2015, 8 of the 19 payers also began covering cfDNA screening in average-risk pregnancies, citing recent CV studies and updated professional guidelines. Most payers attempted, but were unable, to independently assess analytic validity (AV). CONCLUSION Payers utilized the standard evidentiary framework (AV/CV/CU) when evaluating cfDNA screening but varied in their interpretation of the sufficiency of the evidence. Professional guidelines, large CV studies, and decision analytic models regarding health outcomes appeared highly influential in coverage decisions.Genet Med advance online publication 22 September 2016.
Collapse
Affiliation(s)
- Andrew P Dervan
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Patricia A Deverka
- American Institutes for Health Research and Innovation, Chapel Hill, North Carolina, USA
- Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Julia R Trosman
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
- Center for Business Models in Healthcare, Chicago, Illinois, USA
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christine B Weldon
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
- Center for Business Models in Healthcare, Chicago, Illinois, USA
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael P Douglas
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
| | - Kathryn A Phillips
- Department of Clinical Pharmacy, Center for Translational and Policy Research on Personalized Medicine (TRANSPERS), University of California San Francisco, San Francisco, California, USA
- Philip R. Lee Institute for Health Policy, University of California San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
42
|
Haga SB, Moaddeb J, Mills R, Voora D. Assessing feasibility of delivering pharmacogenetic testing in a community pharmacy setting. Pharmacogenomics 2017; 18:327-335. [PMID: 28244804 DOI: 10.2217/pgs-2016-0175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AIM To describe the rationale and design of a study evaluating the delivery of pharmacogenetic (PGx) testing in community pharmacies. Study rationale: Pharmacists have expressed interest in offering PGx testing; however, their lack of knowledge and experience, patients' acceptance and feasibility are unknown in this setting. STUDY DESIGN Through a cluster randomized trial, we will assess pharmacist and patient experiences with delivery of PGx testing as a standalone service or integrated into medication therapy management services. Anticipated results: We anticipate that PGx testing can be delivered in a community pharmacy setting and accepted and valued by patients. CONCLUSION This study is expected to provide valuable evidence about the real-world feasibility and acceptance of a community pharmacist-delivered approach of PGx testing.
Collapse
Affiliation(s)
- Susanne B Haga
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, 304 Research Drive, Box 90141, Durham, NC 27708, USA
| | - Jivan Moaddeb
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, 304 Research Drive, Box 90141, Durham, NC 27708, USA
| | - Rachel Mills
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, 304 Research Drive, Box 90141, Durham, NC 27708, USA
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, 304 Research Drive, Box 90141, Durham, NC 27708, USA
| |
Collapse
|
43
|
Tunis SR, Messner DA. Payer Perspectives on Coverage of Biomarker Testing in Cardiovascular Disease. Clin Chem 2016; 63:258-260. [PMID: 27864388 DOI: 10.1373/clinchem.2016.268136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 10/13/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Sean R Tunis
- Center for Medical Technology Policy, Baltimore, MD
| | | |
Collapse
|
44
|
Hirschtritt ME, Besterman AD, Ross DA. Psychiatric Pharmacogenomics: How Close Are We? Biol Psychiatry 2016; 80:e63-5. [PMID: 27663067 PMCID: PMC5371713 DOI: 10.1016/j.biopsych.2016.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Affiliation(s)
| | - Aaron D Besterman
- Department of Psychiatry, University of California Los Angeles, Los Angeles, California
| | - David A Ross
- Department of Psychiatry, Yale University, New Haven, Connecticut.
| |
Collapse
|
45
|
Moaddeb J, Mills R, Haga SB. Community pharmacists' experience with pharmacogenetic testing. J Am Pharm Assoc (2003) 2016; 55:587-594. [PMID: 26409205 DOI: 10.1331/japha.2015.15017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Appendix 1 Statements of knowledge of correct medication use Appendix 2 Statements of self-efficacy of correct medication use Appendix 3 Statements of skills of correct medication use To characterize the experiences and feasibility of offering pharmacogenetic (PGx) testing in a community pharmacy setting. DESIGN Pharmacists were invited to complete a survey about PGx testing for each patient who was offered testing. If the patient consented, pharmacists were also asked to complete a follow-up survey about the process of returning PGx testing results to patients and follow-up with the prescribing provider. SETTING Community pharmacies in North Carolina from August through November 2014. PARTICIPANTS Pharmacists at five community pharmacies. MAIN OUTCOME MEASURES Patient consent for testing, time to introduce PGx testing initially and communicate results, interpretation of test results, and recommended medication changes. RESULTS Of the 69 patients offered testing, 56 (81%) consented. Pre-test counseling typically lasted 1-5 minutes (81%), and most patients (55%) did not have any questions about the testing. Most pharmacists reported test results to patients by phone (84%), with discussions taking less than 1 minute (48%) or 1-5 minutes (52%). Most pharmacists believed the patients understood their results either very well (54%) or somewhat well (41%). Pharmacists correctly interpreted 47 of the 53 test results (89%). All of the incorrect interpretations were for patients with test results indicating a dosing or drug change (6/19; 32%). Pharmacists reported contacting the ordering physician for four patients to discuss results indicating a dosage or drug change. CONCLUSION The provision of PGx services in a community pharmacy setting appears feasible, requiring little additional time from the pharmacist, and many patients seem interested in PGx testing. Additional training may be necessary to improve test result interpretation, as well as for communication with both patients and ordering physicians.
Collapse
Affiliation(s)
- Jivan Moaddeb
- Clinical Pharmacist, Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Rachel Mills
- Clinical Research Coordinator and Genetic Counselor, Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Susanne B Haga
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
46
|
Messner DA, Al Naber J, Koay P, Cook-Deegan R, Majumder M, Javitt G, Deverka P, Dvoskin R, Bollinger J, Curnutte M, Chandrasekharan S, McGuire A. Barriers to clinical adoption of next generation sequencing: Perspectives of a policy Delphi panel. Appl Transl Genom 2016; 10:19-24. [PMID: 27668172 PMCID: PMC5025465 DOI: 10.1016/j.atg.2016.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/10/2016] [Accepted: 05/23/2016] [Indexed: 04/23/2023]
Abstract
This research aims to inform policymakers by engaging expert stakeholders to identify, prioritize, and deliberate the most important and tractable policy barriers to the clinical adoption of next generation sequencing (NGS). A 4-round Delphi policy study was done with a multi-stakeholder panel of 48 experts. The first 2 rounds of online questionnaires (reported here) assessed the importance and tractability of 28 potential barriers to clinical adoption of NGS across 3 major policy domains: intellectual property, coverage and reimbursement, and FDA regulation. We found that: 1) proprietary variant databases are seen as a key challenge, and a potentially intractable one; 2) payer policies were seen as a frequent barrier, especially a perceived inconsistency in standards for coverage; 3) relative to other challenges considered, FDA regulation was not strongly perceived as a barrier to clinical use of NGS. Overall the results indicate a perceived need for policies to promote data-sharing, and a desire for consistent payer coverage policies that maintain reasonably high standards of evidence for clinical utility, limit testing to that needed for clinical care decisions, and yet also flexibly allow for clinician discretion to use genomic testing in uncertain circumstances of high medical need.
Collapse
Affiliation(s)
- Donna A. Messner
- Center for Medical Technology Policy, 401 East Pratt Street, Suite 631, Baltimore, MD 21207, USA
| | - Jennifer Al Naber
- Center for Medical Technology Policy, 401 East Pratt Street, Suite 631, Baltimore, MD 21207, USA
| | - Pei Koay
- Center for Medical Technology Policy, 401 East Pratt Street, Suite 631, Baltimore, MD 21207, USA
| | | | - Mary Majumder
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Gail Javitt
- Johns Hopkins Berman Institute of Bioethics, 1809 Ashland Avenue, Baltimore, MD 21205, USA
| | - Patricia Deverka
- American Institutes for Research, 1000 Thomas Jefferson Street NW, Washington, DC 20007, USA
| | - Rachel Dvoskin
- Johns Hopkins Berman Institute of Bioethics, 1809 Ashland Avenue, Baltimore, MD 21205, USA
| | - Juli Bollinger
- Johns Hopkins Berman Institute of Bioethics, 1809 Ashland Avenue, Baltimore, MD 21205, USA
| | - Margaret Curnutte
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | - Amy McGuire
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
47
|
Abstract
We examine a recent dispute regarding the Centers for Medicare and Medicaid Services' (CMS) refusal to unconditionally pay for amyloid positron emission tomography (PET) imaging for Medicare beneficiaries being assessed for Alzheimer's disease. CMS will only pay for amyloid PET imaging when patients are enrolled in clinical trials that meet certain criteria. The dispute reflects CMS's willingness in certain circumstances to require effectiveness evidence that differs from the Food and Drug Administration's standard for pre-market approval of a medical intervention and reveals how stakeholders with differing perspectives about evidentiary standards have played a role in attempting to shape the Medicare program's coverage policies.
Collapse
Affiliation(s)
- Karen J Maschke
- a Research Scholar , The Hastings Center , Garrison , New York , USA
| | - Michael K Gusmano
- b Associate Professor of Health Policy , Rutgers University , New Brunswick , New Jersey , USA
| |
Collapse
|
48
|
Delaney SK, Hultner ML, Jacob HJ, Ledbetter DH, McCarthy JJ, Ball M, Beckman KB, Belmont JW, Bloss CS, Christman MF, Cosgrove A, Damiani SA, Danis T, Delledonne M, Dougherty MJ, Dudley JT, Faucett WA, Friedman JR, Haase DH, Hays TS, Heilsberg S, Huber J, Kaminsky L, Ledbetter N, Lee WH, Levin E, Libiger O, Linderman M, Love RL, Magnus DC, Martland A, McClure SL, Megill SE, Messier H, Nussbaum RL, Palaniappan L, Patay BA, Popovich BW, Quackenbush J, Savant MJ, Su MM, Terry SF, Tucker S, Wong WT, Green RC. Toward clinical genomics in everyday medicine: perspectives and recommendations. Expert Rev Mol Diagn 2016; 16:521-32. [PMID: 26810587 PMCID: PMC4841021 DOI: 10.1586/14737159.2016.1146593] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Precision or personalized medicine through clinical genome and exome sequencing has been described by some as a revolution that could transform healthcare delivery, yet it is currently used in only a small fraction of patients, principally for the diagnosis of suspected Mendelian conditions and for targeting cancer treatments. Given the burden of illness in our society, it is of interest to ask how clinical genome and exome sequencing can be constructively integrated more broadly into the routine practice of medicine for the betterment of public health. In November 2014, 46 experts from academia, industry, policy and patient advocacy gathered in a conference sponsored by Illumina, Inc. to discuss this question, share viewpoints and propose recommendations. This perspective summarizes that work and identifies some of the obstacles and opportunities that must be considered in translating advances in genomics more widely into the practice of medicine.
Collapse
Affiliation(s)
- Susan K Delaney
- a Coriell Institute for Medical Research , Camden , NJ , USA
| | - Michael L Hultner
- b Lockheed Martin , Information Systems & Global Solutions , Rockville , MD , USA
| | - Howard J Jacob
- c HudsonAlpha Institute for Biotechnology , Huntsville , AL , USA
| | | | - Jeanette J McCarthy
- e Duke University , Center for Applied Genomics and Precision Medicine , Durham , NC , USA
| | | | - Kenneth B Beckman
- g University of Minnesota , Genomics Center ,, Minneapolis , MN , USA
| | - John W Belmont
- h Baylor College of Medicine , Children's Nutrition Research Center , Houston , TX , USA
| | - Cinnamon S Bloss
- i University of California, San Diego , School of Medicine , La Jolla , CA , USA
| | | | | | - Stephen A Damiani
- k Mission Massimo Foundation , Elsternwick , VIC , Australia .,l Mission Massimo Foundation Inc ., Westlake Village , CA , USA
| | | | | | - Michael J Dougherty
- o The American Society of Human Genetics , Bethesda , MD , USA.,p Department of Pediatrics , University of Colorado School of Medicine , Aurora , CO , USA
| | - Joel T Dudley
- q Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | | | - Jennifer R Friedman
- r University of California, San Diego , Departments of Neurosciences and Pediatrics and Rady Children's Hospital , San Diego , CA , USA
| | | | - Tom S Hays
- t University of Minnesota , Department of Genetics, Cell Biology and Development , Minneapolis , MN , USA
| | | | - Jeff Huber
- u Google Inc ., Mountain View , CA , USA
| | | | | | | | - Elissa Levin
- q Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | | | | | | | - David C Magnus
- y Stanford Center for Biomedical Ethics , Stanford School of Medicine , Stanford , CA , USA
| | | | | | | | - Helen Messier
- ab Healix Health, Ltd , West Vancouver , BC , Canada
| | | | | | | | | | | | | | - Michael M Su
- ai Anthem Blue Cross , Woodland Hills , CA , USA
| | | | - Steven Tucker
- ak Novena Specialist Center , Singapore , Republic of Singapore
| | | | - Robert C Green
- am Division of Genetics, Department of Medicine, Brigham and Women's Hospital , the Broad Institute, Harvard Medical School and Partners Healthcare Personalized Medicine , Boston , MA , USA
| |
Collapse
|
49
|
Crespo-Leiro MG, Stypmann J, Schulz U, Zuckermann A, Mohacsi P, Bara C, Ross H, Parameshwar J, Zakliczyński M, Fiocchi R, Hoefer D, Colvin M, Deng MC, Leprince P, Elashoff B, Yee JP, Vanhaecke J. Clinical usefulness of gene-expression profile to rule out acute rejection after heart transplantation: CARGO II. Eur Heart J 2016; 37:2591-601. [PMID: 26746629 PMCID: PMC5015661 DOI: 10.1093/eurheartj/ehv682] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 11/25/2015] [Indexed: 01/13/2023] Open
Abstract
Aims A non-invasive gene-expression profiling (GEP) test for rejection surveillance of heart transplant recipients originated in the USA. A European-based study, Cardiac Allograft Rejection Gene Expression Observational II Study (CARGO II), was conducted to further clinically validate the GEP test performance. Methods and results Blood samples for GEP testing (AlloMap®, CareDx, Brisbane, CA, USA) were collected during post-transplant surveillance. The reference standard for rejection status was based on histopathology grading of tissue from endomyocardial biopsy. The area under the receiver operating characteristic curve (AUC-ROC), negative (NPVs), and positive predictive values (PPVs) for the GEP scores (range 0–39) were computed. Considering the GEP score of 34 as a cut-off (>6 months post-transplantation), 95.5% (381/399) of GEP tests were true negatives, 4.5% (18/399) were false negatives, 10.2% (6/59) were true positives, and 89.8% (53/59) were false positives. Based on 938 paired biopsies, the GEP test score AUC-ROC for distinguishing ≥3A rejection was 0.70 and 0.69 for ≥2–6 and >6 months post-transplantation, respectively. Depending on the chosen threshold score, the NPV and PPV range from 98.1 to 100% and 2.0 to 4.7%, respectively. Conclusion For ≥2–6 and >6 months post-transplantation, CARGO II GEP score performance (AUC-ROC = 0.70 and 0.69) is similar to the CARGO study results (AUC-ROC = 0.71 and 0.67). The low prevalence of ACR contributes to the high NPV and limited PPV of GEP testing. The choice of threshold score for practical use of GEP testing should consider overall clinical assessment of the patient's baseline risk for rejection.
Collapse
Affiliation(s)
- Maria G Crespo-Leiro
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Universidade da Coruña (UDC), A Coruña, Spain
| | - Jörg Stypmann
- Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Uwe Schulz
- Ruhr University of Bochum, Bad Oeynhausen, Germany
| | | | | | | | | | | | | | | | | | | | - Mario C Deng
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | | | | | - Johan Vanhaecke
- Department of Cardiology, University Hospital Gasthuisberg, Leuven, Belgium
| |
Collapse
|
50
|
Hefti E, Blanco JG. Documenting Pharmacogenomic Testing with CPT Codes. JOURNAL OF AHIMA 2016; 87:56-59. [PMID: 27055343 PMCID: PMC4998735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Pharmacogenomics is the study of how genetics can impact drug response and it aims to maximize therapeutic efficacy while minimizing adverse drug effects in individual patients. The current push for “personalized” approaches to medicine has brought pharmacogenomics to the attention of the healthcare community and the general public. It has been established that the efficacy or safety of certain drugs is impacted in part by the presence of genetic variants and/or other biochemical factors, which may contribute to variable drug response. Consequently, pharmacogenetic testing has been recommended by the FDA to guide the therapy with certain drugs in specific therapeutic settings. As the utilization of pharmacogenetic tests become more ubiquitous in healthcare, there will be a greater need to understand how to document these tests using current procedural terminology (CPT®) codes for the purposes of billing, reimbursement, and recordkeeping. Prior to 2012, most pharmacogenomic tests were considered “molecular pathology” procedures and documented as such. This practice resulted in non-specific documentation that often did not accurately reflect the actual tests performed. The American Medical Association addressed this problem in 2012 and new CPT codes were created to allow for easier, more specific documentation of pharmacogenomic tests. The goal of this article is to provide insight on the status of pharmacogenomic testing, as well as to highlight examples of current CPT codes for tests recommended by the FDA or drug manufacturers to assist in drug therapy. As the utilization of pharmacogenomic testing becomes more widespread, knowledge of how to document these tests will become more valuable.
Collapse
|