1
|
Heath E, Dyson G, Ribeiro JR, Xiu J, Poorman K, Mamdani H, Al-Hallak MN, Shields AF, Elayoubi JA, Winer IS, Cackowski FC, Puckrein GA, Lopes GDL, Jones N, Hauke RJ, Kareff SA, Radovich M, Sledge GW, Spetzler DB, Vidal GA, Marshall JL. Health Disparities among Patients with Cancer Who Received Molecular Testing for Biomarker-Directed Therapy. CANCER RESEARCH COMMUNICATIONS 2024; 4:2598-2609. [PMID: 39172022 PMCID: PMC11450693 DOI: 10.1158/2767-9764.crc-24-0321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
Health disparities present a barrier to successful oncology treatment. The potential for precision oncology to reduce health disparities has not previously been analyzed. We performed a retrospective analysis of 12,627 patients from six major cancer centers whose tumors underwent molecular testing at Caris Life Sciences between 2010 and 2020. Kaplan-Meier and Cox regression were used to describe and analyze overall survival. The molecular and demographic features of the cohort were analyzed by χ2 and ANOVA tests. Black patients composed 25% of the cohort and White patients 63%. Among this molecularly-tested cohort, there were minimal outcome differences based on race, geographic location, or poverty level. When analyzing the interaction of age, race, and sex, racial-based disparities were noted primarily for young non-White women in the study cohort but were more pronounced for men and women of all ages in the broader patient population within the Surveillance, Epidemiology, and End Results database. Mutations in five genes-APC, EGFR, STK11, TP53, and KRAS-were found to affect overall survival among our cohort, and their prevalence varied by race in specific tumor types. Real-world outcomes data in mutation-defined cohorts also provided additional context to previously reported therapeutic response trends. Our study shows that patients who undergo molecular testing display reduced racial health disparities compared with the general population, whereas persistent racial disparities are influenced by age and sex. Genomic-driven racial disparities should be examined at a tumor lineage-specific level. Increased access to molecular testing for all eligible patients may play a role in improving health equity. Significance: This study is the largest of its kind to analyze health disparities and genomic features among a diverse multiinstitutional cohort of patients who underwent molecular testing. Continuing to increase awareness of and access to molecular testing approaches may help to reduce cancer health disparities and improve outcomes for all patients.
Collapse
Affiliation(s)
- Elisabeth Heath
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Gregory Dyson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | | | | | | | - Hirva Mamdani
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Mohammed N. Al-Hallak
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Anthony F. Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Jailan A. Elayoubi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Ira S. Winer
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Frank C. Cackowski
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Gary A. Puckrein
- National Minority Quality Forum, Washington, District of Columbia.
| | - Gilberto de Lima Lopes
- Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.
| | - Nathaniel Jones
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.
| | | | - Samuel A. Kareff
- Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.
| | | | | | | | - Gregory A. Vidal
- West Cancer Center and Research Institute, Germantown, Tennessee.
| | - John L. Marshall
- Division of Hematology and Oncology, MedStar Health/Lombardi Comprehensive Cancer Center Georgetown University, Washington, District of Columbia.
| |
Collapse
|
2
|
Ferreira-Gonzalez A, Ko G, Fusco N, Stewart F, Kistler K, Appukkuttan S, Hocum B, Allen SM, Babajanyan S. Barriers and facilitators to next-generation sequencing use in United States oncology settings: a systematic review. Future Oncol 2024; 20:2765-2777. [PMID: 39316553 DOI: 10.1080/14796694.2024.2390821] [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: 11/21/2023] [Accepted: 08/07/2024] [Indexed: 09/26/2024] Open
Abstract
Aim: Next-generation sequencing (NGS) of solid tumors can inform treatment decisions; however, uptake remains low. This objective of this systematic review was to identify barriers to and facilitators of NGS in US oncology settings.Materials & methods: Embase and MEDLINE were searched in March 2023 for articles published from 2012 to 2023 on barriers and facilitators of NGS adoption for solid tumors. Surveys, interviews and observational studies were eligible. Studies on genetic testing for hereditary cancers and non-US studies were excluded. The Motheral scale, Joanna Briggs Institute critical appraisal checklist and McGill Mixed Methods Appraisal Tool were used to assess study quality. Data were synthesized narratively.Results: Twenty-one studies were included. Study participants were clinicians, payers and administrators. Key barriers included complex reimbursement processes and uncertainties around clinical utility. Including recommendations for NGS in clinical practice guidelines was a key facilitator, although insurance policies were often more restrictive than guideline recommendations.Conclusion: Uptake of NGS is increasing but barriers remain. Changes to the current reimbursement frameworks are needed to increase access to NGS. The impact of implementing the 2018 National Coverage Determination, which allows access to NGS for all Medicare beneficiaries with advanced cancer, is not yet evident in the published literature.
Collapse
Affiliation(s)
| | - Gilbert Ko
- Health Economics and Outcomes Research, Bayer Healthcare US, L.L.C., Whippany, NJ 07981, USA
| | - Nicole Fusco
- Evidence Generation and Value Communications, Cencora, Conshohocken, PA 19427, USA
| | - Fiona Stewart
- Evidence Generation and Value Communications, Cencora, Conshohocken, PA 19427, USA
| | - Kristin Kistler
- Evidence Generation and Value Communications, Cencora, Conshohocken, PA 19427, USA
| | - Sreevalsa Appukkuttan
- Health Economics and Outcomes Research, Bayer Healthcare US, L.L.C., Whippany, NJ 07981, USA
| | - Brian Hocum
- Health Economics and Outcomes Research, Bayer Healthcare US, L.L.C., Whippany, NJ 07981, USA
| | - Stefan M Allen
- Health Economics and Outcomes Research, Bayer Healthcare US, L.L.C., Whippany, NJ 07981, USA
| | | |
Collapse
|
3
|
Liu Z, Zhao J, Cui K, Guo H, Li Z, Zhou Z. Detection accuracy and clinical applications of DP-TOF mass spectrometry. J Int Med Res 2024; 52:3000605241255568. [PMID: 38819085 PMCID: PMC11143829 DOI: 10.1177/03000605241255568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE Matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) is currently used in clinical microbiology laboratories. This study aimed to determine whether dual-polarity time-of-flight mass spectrometry (DP-TOF MS) could be applied to clinical nucleotide detection. METHODS This prospective study included 40 healthy individuals and 110 patients diagnosed with cardiovascular diseases. We used DP-TOF MS and Sanger sequencing to evaluate 17 loci across 11 genes associated with cardiovascular drug responses. In addition, we used DP-TOF MS to test 998 retrospectively collected clinical DNA samples with known results. RESULTS A, T, and G nucleotide detection by DP-TOF MS and Sanger sequencing revealed 100% concordance, whereas the C nucleotide concordance was 99.86%. Genotyping based on the results of the two methods showed 99.96% concordance. Regarding clinical applications, DP-TOF MS yielded a 99.91% concordance rate for known loci. The minimum detection limit for DNA was 0.4 ng; the inter-assay and intra-assay precision rates were both 100%. Anti-interference analysis showed that aerosol contamination greater than 1013 copies/µL in the laboratory environment could influence the results of DP-TOF MS. CONCLUSIONS The DP-TOF MS platform displayed good detection performance, as demonstrated by its 99.96% concordance rate with Sanger sequencing. Thus, it may be applied to clinical nucleotide detection.
Collapse
Affiliation(s)
- Zhaohui Liu
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Center of Laboratory Medicine, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, China
| | - Juan Zhao
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Cui
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huimin Guo
- Zhejiang Digena Diagnosis Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Zhikai Li
- Zhejiang Digena Diagnosis Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
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: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
5
|
Lazure P, Sireci A, Subbiah V, Murray S, Grohé C, Sherman SI, Kelly E, Bubach P, Péloquin S. Challenges in diagnosis and biomarker testing for RET-altered lung and thyroid cancer care: an international mixed-method study. BMC MEDICAL EDUCATION 2023; 23:410. [PMID: 37277734 DOI: 10.1186/s12909-023-04396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND The introduction of new targeted therapies for RET-altered lung and thyroid cancers (LC/TC) has impacted pathologists' practice by making genomic testing more relevant. Variations in health systems and treatment access result in distinct clinical challenges and barriers. This study aimed to assess practice gaps and challenges experienced by pathologists involved in the diagnosis of RET-altered LC/TC, including biomarker testing, to inform educational solutions. METHODS Pathologists in Germany, Japan, the UK, and US participated in this ethics-approved mixed-methods study, which included interviews and surveys (data collected January-March 2020). Qualitative data was thematically analysed, quantitative data was analysed with chi-square and Kruskal-Wallis H-tests, and both were triangulated. RESULTS A total of 107 pathologists took part in this study. Knowledge gaps were reported regarding genomic testing for LC/TC in Japan (79/60%), the UK (73/66%), and the US (53/30%). Skill gaps were reported when selecting genomic biomarker tests to diagnose TC in Japan (79%), the UK (73%) and US (57%) and when performing specific biomarker tests, especially in Japan (82% for RET) and in the UK (75% for RET). Japanese participants (80%) reported uncertainty about what information to share with the multidisciplinary team to ensure optimal patient-centered care. At the time of data collection, pathologists in Japan faced access barriers to using RET biomarker tests: only 28% agreed that there are relevant RET genomic biomarker tests available in Japan, versus 67% to 90% in other countries. CONCLUSIONS This study identified areas where pathologists need additional continuing professional development opportunities to enhance their competencies and better support delivery of care to patients with RET-altered lung or thyroid tumours. Addressing identified gaps and improving competencies of pathologists in this field should be emphasised in continuing medical education curricula and through quality improvement initiatives. Strategies deployed on an institutional and health system level should aim to improve interprofessional communication and genetic biomarker testing expertise.
Collapse
Affiliation(s)
- Patrice Lazure
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada.
| | | | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suzanne Murray
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada
| | | | - Steven I Sherman
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sophie Péloquin
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 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
|
7
|
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: 0.5] [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
|
8
|
Sadigh G, Goeckner HG, Kazerooni EA, Johnson BE, Smith RA, Adams DV, Carlos RC. State legislative trends related to biomarker testing. Cancer 2022; 128:2865-2870. [PMID: 35607821 DOI: 10.1002/cncr.34271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/13/2022] [Accepted: 04/25/2022] [Indexed: 11/06/2022]
Abstract
Comprehensive biomarker testing has become the standard of care for informing the choice of the most appropriate targeted therapy for many patients with advanced cancer. Despite evidence demonstrating the need for comprehensive biomarker testing to enable the selection of appropriate targeted therapies and immunotherapy, the incorporation of biomarker testing into clinical practice lags behind recommendations in National Comprehensive Cancer Network guidelines. Coverage policy differences across insurance health plans have limited the accessibility of comprehensive biomarker testing largely to patients whose insurance covers the recommended testing or those who can pay for the testing, and this has contributed to health disparities. Furthermore, even when insurance coverage exists for recommended biomarker testing, patients may incur burdensome out-of-pocket costs depending on their insurance plan benefits, which may also create barriers to testing. Prior authorization for biomarker testing for some patients can add an administrative burden and may delay testing and thus treatment if it is not done in a timely manner. Recently, three states (Illinois, Louisiana, and California) passed laws designed to improve access to biomarker testing at the state level. However, there is variability among these laws in terms of the population affected, the stage of cancer, and whether the coverage of testing is mandated, or the legislation addresses only prior authorization. Advocacy efforts by patient advocates, health care professionals, and professional societies are imperative at the state level to further improve coverage for and access to appropriate biomarker testing.
Collapse
Affiliation(s)
- Gelareh Sadigh
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hilary Gee Goeckner
- American Cancer Society Cancer Action Network, Inc, Washington, District of Columbia, USA
| | - Ella A Kazerooni
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bruce E Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Robert A Smith
- Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| | - Devon V Adams
- American Cancer Society Cancer Action Network, Inc, Washington, District of Columbia, USA
| | - Ruth C Carlos
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Acharya M, Zorn KK, Simonson ME, Bimali M, Moore GW, Peng C, Martin BC. Statewide trends and factors associated with genetic testing for hereditary cancer risk in Arkansas 2013-2018. Hered Cancer Clin Pract 2022; 20:19. [PMID: 35606835 PMCID: PMC9128197 DOI: 10.1186/s13053-022-00226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early identification of hereditary cancer risk would save lives, but genetic testing (GT) has been inadequate. We assessed i) trends for hereditary breast and ovarian cancer (HBOC), Lynch syndrome, and other GT and ii) factors associated with receipt of GT. METHODS We used data from the Arkansas All-Payer Claims Database from January 2013 through June 2018 (commercial, Medicaid), December 2017 (state employee), or December 2016 (Medicare) and identified enrollees with ≥1 month of enrollment. Using Current Procedural Terminology (CPT-4) codes, rates for GT were calculated per 100,000 person-quarters and time series regressions estimated. Second, GT and covariate information for enrollees with 24 months of continuous enrollment were used to estimate separate logistic regression models for each GT category. RESULTS Among 2,520,575 unique enrollees, HBOC testing rates were 2.2 (Medicaid), 22.0 (commercial), 40.4 (state employee), and 13.1(Medicare) per 100,000 person-quarters and increased linearly across all plans. Older age (OR=1.24; 95%CI 1.20 - 1.28), female sex (OR=18.91; 95%CI 13.01 - 28.86), higher comorbidity burden (OR=1.08; 95%CI 1.05 - 1.12), mental disorders (OR=1.53; 95%CI 1.15 - 2.00), and state employee coverage (OR=1.65; 95%CI 1.37 - 1.97) were positively associated with HBOC testing. Less than 1 of 10,000 enrollees received Lynch syndrome testing, while < 5 of 10,000 received HBOC testing. CONCLUSION GT rates for hereditary cancer syndromes have increased in Arkansas but remain low. Receipt of GT was explained with high discrimination by sex and plan type. IMPACT Expansion of GT for hereditary cancer risk in Arkansas is needed to identify high-risk individuals who could benefit from risk-reduction strategies.
Collapse
Affiliation(s)
- Mahip Acharya
- Division of Pharmaceutical Evaluation and Policy, Department of Pharmacy Practice, University of Arkansas for Medical Sciences College of Pharmacy, Education II Building 6253, 4301 W. Markham Street, slot 522, Little Rock, AR-72205, United States
| | - Kristin K Zorn
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences College of Medicine, Little Rock, AR, United States
| | - Melinda E Simonson
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences College of Medicine, Little Rock, AR, United States
| | - Milan Bimali
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Gary W Moore
- Division of Pharmaceutical Evaluation and Policy, Department of Pharmacy Practice, University of Arkansas for Medical Sciences College of Pharmacy, Education II Building 6253, 4301 W. Markham Street, slot 522, Little Rock, AR-72205, United States
| | - Cheng Peng
- Division of Pharmaceutical Evaluation and Policy, Department of Pharmacy Practice, University of Arkansas for Medical Sciences College of Pharmacy, Education II Building 6253, 4301 W. Markham Street, slot 522, Little Rock, AR-72205, United States
| | - Bradley C Martin
- Division of Pharmaceutical Evaluation and Policy, Department of Pharmacy Practice, University of Arkansas for Medical Sciences College of Pharmacy, Education II Building 6253, 4301 W. Markham Street, slot 522, Little Rock, AR-72205, United States.
| |
Collapse
|
10
|
Wong WB, Anina D, Lin CW, Adams DV. Alignment of health plan coverage policies for somatic multigene panel testing with clinical guidelines in select solid tumors. Per Med 2022; 19:171-180. [PMID: 35118882 DOI: 10.2217/pme-2021-0174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Aim: Commercial plan coverage policies for multigene panel tests may vary and could result in geographic variation in coverage due to the fragmented nature of the commercial insurance market. This study aimed to characterize the alignment of multigene panel tests coverage policies to that of clinical guidelines, overall and by state. Materials & methods: We reviewed NCCN Guidelines® for four tumors. Public coverage policies were identified via web search. Payer policies included those with the largest or second largest number of commercial lives in each state. Policies were classified as 'more restrictive' or 'consistent' with the guidelines. Results: Of 38 plans/policies reviewed, 71% were classified as 'more restrictive' than the guidelines, with variation in the number of commercial lives by state. Among these, 52% restricted on panel size and 63% restricted in all or select tumors. Conclusion: Most coverage policies were more restrictive. Clinical guideline clarity and state policies may improve alignment to guidelines and geographic variations.
Collapse
Affiliation(s)
- William B Wong
- Health Policy and Systems Research, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Chia-Wei Lin
- Evidence for Access, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Devon V Adams
- Policy and Legislative Support, American Cancer Society Cancer Action Network, Washington, DC 20005, USA
| |
Collapse
|
11
|
Sheinson DM, Wong WB, Meyer CS, Stergiopoulos S, Lofgren KT, Flores C, Adams DV, Fleury ME. Trends in Use of Next-Generation Sequencing in Patients With Solid Tumors by Race and Ethnicity After Implementation of the Medicare National Coverage Determination. JAMA Netw Open 2021; 4:e2138219. [PMID: 34882180 PMCID: PMC8662372 DOI: 10.1001/jamanetworkopen.2021.38219] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/12/2021] [Indexed: 12/23/2022] Open
Abstract
Importance In March 2018, Medicare issued a national coverage determination (NCD) for next-generation sequencing (NGS) to facilitate access to NGS testing among Medicare beneficiaries. It is unknown whether the NCD affected health equity issues for Medicare beneficiaries and the overall population. Objective To examine the association between the Medicare NCD and NGS use by insurance types and race and ethnicity. Design, Setting, and Participants A retrospective cohort analysis was conducted using electronic health record data derived from a real-world database. Data originated from approximately 280 cancer clinics (approximately 800 sites of care) in the US. Patients with advanced non-small cell lung cancer (aNSCLC), metastatic colorectal cancer (mCRC), metastatic breast cancer (mBC), or advanced melanoma diagnosed from January 1, 2011, through March 31, 2020, were included. Exposure Pre- vs post-NCD period. Main Outcomes and Measures Patients were classified by insurance type and race and ethnicity to examine patterns in NGS testing less than or equal to 60 days after diagnosis. Difference-in-differences models examined changes in average NGS testing in the pre- and post-NCD periods by race and ethnicity, and interrupted time-series analysis examined whether trends over time varied by insurance type and race and ethnicity. Results Among 92 687 patients with aNSCLC, mCRC, mBC, or advanced melanoma, mean (SD) age was 66.6 (11.2) years, 51 582 (55.7%) were women, and 63 864 (68.9%) were Medicare beneficiaries. The largest racial and ethnic categories according to the database used and further classification were Black or African American (8605 [9.3%]) and non-Hispanic White (59 806 [64.5%]). Compared with Medicare beneficiaries, changes in pre- to post-NCD NGS testing trends were similar in commercially insured patients (odds ratio [OR], 1.03; 95% CI, 0.98-1.08; P = .25). Pre- to post-NCD NGS testing trends increased at a slower rate among patients in assistance programs (OR, 0.93; 95% CI, 0.87-0.99; P = .03) compared with Medicare beneficiaries. The rate of increase for patients receiving Medicaid was not statistically significantly different compared with those receiving Medicare (OR, 0.92; 95% CI, 0.84-1.01; P = .07). The NCD was not associated with statistically significant changes in NGS use trends by racial and ethnic groups within Medicare beneficiaries alone or across all insurance types. Compared with non-Hispanic White individuals, increases in average NGS use from the pre-NCD to post-NCD period were 14% lower (OR, 0.86; 95% CI, 0.74-0.99; P = .04) among African American and 23% lower (OR, 0.77; 95% CI, 0.62-0.96; P = .02) among Hispanic/Latino individuals; increases among Asian individuals and those with other races and ethnicities were similar. Conclusions and Relevance The findings of this study suggest that expansion of Medicare-covered benefits may not occur equally across insurance types, thereby further widening or maintaining disparities in NGS testing. Additional efforts beyond coverage policies are needed to ensure equitable access to the benefits of precision medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Devon V. Adams
- American Cancer Society Cancer Action Network, Washington, DC
| | - Mark E. Fleury
- American Cancer Society Cancer Action Network, Washington, DC
| |
Collapse
|
12
|
Elchynski AL, Cicali EJ, Ferrer Del Busto MC, Hamilton A, Chang KL, Schmidt SO, Weiner B, Davis R, Estores D, Max Smith D, Wiisanen K, Johnson JA, Cavallari LH. Determining the potential clinical value of panel-based pharmacogenetic testing in patients with chronic pain or gastroesophageal reflux disease. THE PHARMACOGENOMICS JOURNAL 2021; 21:657-663. [PMID: 34075203 PMCID: PMC8605985 DOI: 10.1038/s41397-021-00244-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/29/2021] [Accepted: 05/19/2021] [Indexed: 12/31/2022]
Abstract
We aimed to determine the potential value of panel-based pharmacogenetic (PGx) testing in patients with chronic pain or gastroesophageal reflux disease (GERD) who underwent single-gene PGx testing to guide opioid or proton pump inhibitor (PPI) therapy, respectively. Of 448 patients included (chronic pain, n = 337; GERD, n = 111), mean age was 57 years, 68% were female, and 73% were white. Excluding opiates for the pain cohort and PPIs for the GERD cohort, 76.6% of patients with pain and 71.2% with GERD were prescribed at least one additional medication with a high level of PGx evidence, most commonly ondansetron or selective serotonin reuptake inhibitors. The most common genes that could inform PGx drug prescribing were CYP2C19, CYP2D6, CYP2C9, and SLCO1B1. Our findings suggest that patients with chronic pain or GERD are commonly prescribed drugs with a high level of evidence for a PGx-guided approach, supporting panel-based testing in these populations.
Collapse
Affiliation(s)
- Amanda L Elchynski
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Emily J Cicali
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Maria C Ferrer Del Busto
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Alessandra Hamilton
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ku-Lang Chang
- Department of Community Health and Family Medicine, College of Medicine University of Florida, Gainesville, FL, USA
| | - Siegfried O Schmidt
- Department of Community Health and Family Medicine, College of Medicine University of Florida, Gainesville, FL, USA
| | - Brian Weiner
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, FL, USA
- Charles E. Schmidt School of Medicine, Florida Atlantic University, Boca Raton, FL, USA
- Digestive Disease Institute at the Cleveland Clinic, Weston, FL, USA
| | - Richard Davis
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, FL, USA
| | - David Estores
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, FL, USA
| | - D Max Smith
- MedStar Health, Columbia, MD, USA
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Kristin Wiisanen
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Larisa H Cavallari
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA.
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
13
|
Sheinson DM, Wong WB, Flores C, Ogale S, Gross CP. Association Between Medicare's National Coverage Determination and Utilization of Next-Generation Sequencing. JCO Oncol Pract 2021; 17:e1774-e1784. [PMID: 34043456 PMCID: PMC8600504 DOI: 10.1200/op.20.01023] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/18/2021] [Accepted: 04/26/2021] [Indexed: 12/30/2022] Open
Abstract
PURPOSE In 2018, Medicare issued a national coverage determination (NCD) providing reimbursement for next-generation sequencing (NGS) tests for beneficiaries with advanced or metastatic cancer and no previous NGS testing. We examined the association between NCD implementation and NGS utilization trends in Medicare beneficiaries versus commercially insured patients. METHODS This was a retrospective study of patients with advanced non-small-cell lung cancer (aNSCLC), metastatic colorectal cancer (mCRC), metastatic breast cancer (mBC), or advanced melanoma with a de novo or recurrent advanced diagnosis from January 1, 2011, through December 30, 2019, using a nationwide US electronic health record-derived deidentified database. Patients were classified by insurance and by advanced diagnosis date. NGS testing was assessed by receipt of first NGS test result ≤ 60 days of advanced diagnosis. Interrupted time series analysis assessed NGS utilization pre- and post-NCD effective date by insurance type. RESULTS The utilization and repeat NGS testing analysis included 70,290 and 4,295 patients, respectively. Use of NGS rose from < 1% in 2011 to > 45% in Q4 2019 in aNSCLC while remaining < 20% in mBC and advanced melanoma. Among patients with aNSCLC, mCRC, or mBC, NGS testing increased post-NCD versus pre-NCD (P < .05). There was no significant difference in trends pre- and post-NCD between Medicare beneficiaries and commercially insured patients in any tumor. Repeat NGS testing was similar before the NCD (Medicare v commercial: 24.8% v 28.5%). Post-NCD, fewer Medicare beneficiaries had repeat NGS testing (27.7% v 36.0%; P < .01). CONCLUSION Trends in NGS utilization significantly changed post-NCD, although the magnitude of change was not significantly different by insurance type, indicating private insurers may also be incorporating NCD guidance. Implementation of the NCD may have limited use of repeat NGS testing in Medicare beneficiaries.
Collapse
Affiliation(s)
| | | | | | | | - Cary P. Gross
- Cancer Outcomes Public Policy and Effectiveness Research (COPPER) Center, Yale Cancer Center, New Haven, CT
| |
Collapse
|
14
|
Gamble CR, Huang Y, Wright JD, Hou JY. Precision medicine testing in ovarian cancer: The growing inequity between patients with commercial vs medicaid insurance. Gynecol Oncol 2021; 162:18-23. [PMID: 33958212 DOI: 10.1016/j.ygyno.2021.04.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/20/2021] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Precision medicine technologies have significant impact in the care of patients with ovarian cancer. Compared to affluent patients, socioeconomically vulnerable patients are less likely to have access to this testing. There is little data that demonstrate this inequity over time. METHODS We used the IBM Truven Health MarketScan Research Database to identify patients in the United States who underwent surgery for ovarian cancer between 2011 and 2017. The presence of claims for precision medicine testing within six months of surgery was assessed for each patient. Precision medicine testing included both molecular genetic testing (BRCA limited or full sequencing, somatic and germline testing) as well as ancillary pathology tests (immunohistochemistry, microsatellite instability). Demographic data was extracted. RESULTS We identified 27,181 patients who met eligibility. Of these, 88.6% had commercial insurance, and 11.4% had Medicaid. While the proportion of patients who underwent precision medicine testing increased over time for both cohorts (47.0% to 66.6% for commercially insured, 41.4% to 57.6% for Medicaid insured, p < 0.0001), the inequity in testing rates widened (5.6% disparity to 9.0%, p < 0.0001). This was driven by growing inequity in germline and somatic genetic testing (7.6% disparity to 21.3%, p < 0.0001). CONCLUSIONS There is widening inequity in precision medicine testing rates between commercially insured and Medicaid insured poate patients with ovarian cancer.
Collapse
Affiliation(s)
- Charlotte R Gamble
- Columbia University College of Physicians and Surgeons, United States of America; New York Presbyterian Hospital, United States of America.
| | - Yongmei Huang
- Columbia University College of Physicians and Surgeons, United States of America; Joseph L. Mailman School of Public Health, Columbia University, United States of America
| | - Jason D Wright
- Columbia University College of Physicians and Surgeons, United States of America; Herbert Irving Comprehensive Cancer Center, United States of America; New York Presbyterian Hospital, United States of America
| | - June Y Hou
- Columbia University College of Physicians and Surgeons, United States of America; Herbert Irving Comprehensive Cancer Center, United States of America; New York Presbyterian Hospital, United States of America
| |
Collapse
|
15
|
Feofanova EV, Zhang GQ, Lhatoo S, Metcalf GA, Boerwinkle E, Venner E. The Implementation Science for Genomic Health Translation (INSIGHT) Study in Epilepsy: Protocol for a Learning Health Care System. JMIR Res Protoc 2021; 10:e25576. [PMID: 33769305 PMCID: PMC8088873 DOI: 10.2196/25576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/11/2021] [Accepted: 02/25/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Genomic medicine is poised to improve care for common complex diseases such as epilepsy, but additional clinical informatics and implementation science research is needed for it to become a part of the standard of care. Epilepsy is an exemplary complex neurological disorder for which DNA diagnostics have shown to be advantageous for patient care. OBJECTIVE We designed the Implementation Science for Genomic Health Translation (INSIGHT) study to leverage the fact that both the clinic and testing laboratory control the development and customization of their respective electronic health records and clinical reporting platforms. Through INSIGHT, we can rapidly prototype and benchmark novel approaches to incorporating clinical genomics into patient care. Of particular interest are clinical decision support tools that take advantage of domain knowledge from clinical genomics and can be rapidly adjusted based on feedback from clinicians. METHODS Building on previously developed evidence and infrastructure components, our model includes the following: establishment of an intervention-ready genomic knowledge base for patient care, creation of a health informatics platform and linking it to a clinical genomics reporting system, and scaling and evaluation of INSIGHT following established implementation science principles. RESULTS INSIGHT was approved by the Institutional Review Board at the University of Texas Health Science Center at Houston on May 15, 2020, and is designed as a 2-year proof-of-concept study beginning in December 2021. By design, 120 patients from the Texas Comprehensive Epilepsy Program are to be enrolled to test the INSIGHT workflow. Initial results are expected in the first half of 2023. CONCLUSIONS INSIGHT's domain-specific, practical but generalizable approach may help catalyze a pathway to accelerate translation of genomic knowledge into impactful interventions in patient care. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/25576.
Collapse
Affiliation(s)
- Elena Valeryevna Feofanova
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Guo-Qiang Zhang
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, United States
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Texas Institute for Restorative Neurotechnologies, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Samden Lhatoo
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Texas Institute for Restorative Neurotechnologies, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ginger A Metcalf
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, United States
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, United States
| | - Eric Venner
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
16
|
Scott A, Martin DM. Development and implementation of an electronic medical record module to track genetic testing results. Genet Med 2021; 23:972-975. [PMID: 33500566 DOI: 10.1038/s41436-020-01057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/24/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Genetic testing and results return pose many challenges, even in the era of electronic medical records. Whether results are positive or negative, genetic testing and return of results necessitate patient follow-up, referrals, and coordination between providers. Genetic evaluations typically utilize a variety of testing modalities with differing timetables and/or avenues to return. Therefore, genetic information requires a secondary, unified mechanism for storing and tracking results and communication to facilitate patient care. METHODS We developed an electronic medical record (EMR) episodes-based module called Pediatric Genetic Tracking to provide a centralized summary of patient tracking information in a single-institution pediatric genetics setting. RESULTS We created episodes for 6,133 patients evaluated in our division over a 3-year period. They highlighted clinical information for 1,901 different diagnoses and 547 genetic tests, and the involvement of 9 providers, 7 genetic counselors, 61 trainees, and 15 students using two modes of follow-up. CONCLUSION This Pediatric Genetic Tracking episodes system serves as a "one-stop shop" living document for updated patient genetic information and can be easily expanded to include variant content for broader population level sharing or analysis. These episodes-based modules facilitate communication to support timely and accurate return of genetic test results and follow-up.
Collapse
Affiliation(s)
- Anthony Scott
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| | - Donna M Martin
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.,Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| |
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: 2.3] [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
|
Burnett-Hartman AN, Udaltsova N, Kushi LH, Neslund-Dudas C, Rahm AK, Pawloski PA, Corley DA, Knerr S, Feigelson HS, Hunter JE, Tabano DC, Epstein MM, Honda SA, Ter-Minassian M, Lynch JA, Lu CY. Clinical Molecular Marker Testing Data Capture to Promote Precision Medicine Research Within the Cancer Research Network. JCO Clin Cancer Inform 2020; 3:1-10. [PMID: 31487201 DOI: 10.1200/cci.19.00026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE To evaluate health care systems for the availability of population-level data on the frequency of use and results of clinical molecular marker tests to inform precision cancer care. METHODS We assessed cancer-related molecular marker test data availability across 12 US health care systems in the Cancer Research Network. Overall, these systems provide care to a diverse population of more than 12 million people in the United States. We performed qualitative analyses of test data availability for five blood-based protein, nine germline, and 14 tissue-based tumor marker tests in each health care system's electronic health record and tumor registry using key informants, test code lists, and manual review of data types and output. We then performed quantitative analyses to estimate the proportion of patients with cancer with test utilization data and results for specific molecular marker tests. RESULTS Health systems were able to systematically capture population-level data on all five blood protein markers, six of 14 tissue-based tumor markers, and none of the nine germline markers. Successful, systematic data capture was achievable for tests with electronic data feeds for test results (blood protein markers) or through prior manual abstraction by tumor registrars (select tumor-based markers). For test results stored in scanned image files (particularly germline and tumor marker tests), information on which test was performed and test results was not readily accessible in an electronic format. CONCLUSION Even in health care systems with sophisticated electronic health records, there were few codified data elements available for evaluating precision cancer medicine test use and results at the population level. Health care organizations should establish standards for electronic reporting of precision medicine tests to expedite cancer research and facilitate the implementation of precision medicine approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sarah Knerr
- University of Washington and Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | | | | | - David C Tabano
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO
| | - Mara M Epstein
- University of Massachusetts Medical School, Worcester, MA
| | | | | | - Julie A Lynch
- Department of Veterans Affairs Salt Lake City Health System, Salt Lake City, UT
| | - Christine Y Lu
- Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| |
Collapse
|
19
|
Thoreson E, Murphy L, Lemons J, Farach L, Wilson K, Woodson A, Wagner C. Impacts of genesurance considerations on genetic counselors' practice and attitudes. J Genet Couns 2020; 29:1210-1220. [PMID: 32432815 DOI: 10.1002/jgc4.1291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 01/19/2023]
Abstract
Genesurance counseling has been identified as an integral part of many genetic counseling sessions, but little is known about the workflow impacts and genetic counselor perceptions of genesurance-related tasks. In this study, we aimed to characterize how insurance and billing considerations for genetic testing are being incorporated into genetic counselors' practice in the United States, as well as describe current attitudes and challenges associated with their integration. An electronic survey was sent by email to members of the National Society of Genetic Counselors (NSGC). A total of 325 American Board of Genetic Counselors-certified genetic counselors who provide direct patient care in the United States for at least 50% of their time were included in data analysis. Results showed that the frequency and timing of various insurance- and billing-related tasks were not consistent among respondents, even those practicing in similar settings. Inadequate training to complete tasks was reported by 64% of respondents, and 47% reported a lack of resources from their employer and/or institution to complete genesurance tasks. Additionally, only 38% of respondents agreed that insurance- and billing-related tasks were within the scope of the genetic counseling practice, and there was little consensus on who respondents believe is the most appropriate person to complete these tasks. When asked how genesurance considerations affected job satisfaction, 85% of respondents reported a negative impact. This study found an inconsistent genesurance workflow among genetic counselors practicing in the United States, a lack of consensus on who should be responsible for genesurance tasks, several challenges associated with completing these tasks, and identifies genesurance considerations as potential risk factors for genetic counselor burnout.
Collapse
Affiliation(s)
- Emily Thoreson
- University of Texas Genetic Counseling Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lauren Murphy
- University of Texas Genetic Counseling Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jennifer Lemons
- University of Texas Genetic Counseling Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Pediatrics, Division of Medical Genetics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Laura Farach
- University of Texas Genetic Counseling Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Pediatrics, Division of Medical Genetics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kate Wilson
- Advanced Diagnostics, Quest Diagnostics Laboratory, Atlanta, GA, USA
| | - Ashley Woodson
- University of Texas Genetic Counseling Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Clinical Cancer Genetics Program, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chelsea Wagner
- University of Texas Genetic Counseling Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
20
|
Trosman JR, Douglas MP, Liang SY, Weldon CB, Kurian AW, Kelley RK, Phillips KA. Insights From a Temporal Assessment of Increases in US Private Payer Coverage of Tumor Sequencing From 2015 to 2019. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:551-558. [PMID: 32389219 PMCID: PMC7217867 DOI: 10.1016/j.jval.2020.01.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/09/2019] [Accepted: 01/08/2020] [Indexed: 05/12/2023]
Abstract
OBJECTIVES To examine the temporal trajectory of insurance coverage for next-generation tumor sequencing (sequencing) by private US payers, describe the characteristics of coverage adopters and nonadopters, and explore adoption trends relative to the Centers for Medicare and Medicaid Services' National Coverage Determination (CMS NCD) for sequencing. METHODS We identified payers with positive coverage (adopters) or negative coverage (nonadopters) of sequencing on or before April 1, 2019, and abstracted their characteristics including size, membership in the BlueCross BlueShield Association, and whether they used a third-party policy. Using descriptive statistics, payer characteristics were compared between adopters and nonadopters and between pre-NCD and post-NCD adopters. An adoption timeline was constructed. RESULTS Sixty-nine payers had a sequencing policy. Positive coverage started November 30, 2015, with 1 payer and increased to 33 (48%) as of April 1, 2019. Adopters were less likely to be BlueCross BlueShield members (P < .05) and more likely to use a third-party policy (P < .001). Fifty-eight percent of adopters were small payers. Among adopters, 52% initiated coverage pre-NCD over a 25-month period and 48% post-NCD over 17 months. CONCLUSIONS We found an increase, but continued variability, in coverage over 3.5 years. Temporal analyses revealed important trends: the possible contribution of the CMS NCD to a faster pace of coverage adoption, the interdependence in coverage timing among BlueCross BlueShield members, the impact of using a third-party policy on coverage timing, and the importance of small payers in early adoption. Our study is a step toward systematic temporal research of coverage for precision medicine, which will inform policy and affordability assessments.
Collapse
Affiliation(s)
- Julia R Trosman
- Center for Translational and Policy Research on Personalized Medicine, University of California at San Francisco, San Francisco, CA, USA; Center for Business Models in Healthcare, Chicago, IL, USA.
| | - Michael P Douglas
- Center for Translational and Policy Research on Personalized Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Su-Ying Liang
- Palo Alto Medical Foundation Research Institute, Palo Alto, CA, USA
| | - Christine B Weldon
- Center for Translational and Policy Research on Personalized Medicine, University of California at San Francisco, San Francisco, CA, USA; Center for Business Models in Healthcare, Chicago, IL, USA
| | - Allison W Kurian
- Departments of Medicine & of Health Research & Policy, Stanford University, Palo Alto, CA, USA
| | - Robin K Kelley
- Philip R. Lee Institute for Health Policy, University of California, San Francisco, San Francisco, CA, USA
| | - Kathryn A Phillips
- Center for Translational and Policy Research on Personalized Medicine, University of California at San Francisco, San Francisco, CA, USA; Philip R. Lee Institute for Health Policy, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
21
|
Deverka PA, Douglas MP, Phillips KA. Use of Real-World Evidence in US Payer Coverage Decision-Making for Next-Generation Sequencing-Based Tests: Challenges, Opportunities, and Potential Solutions. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:540-550. [PMID: 32389218 PMCID: PMC7219085 DOI: 10.1016/j.jval.2020.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/26/2020] [Accepted: 02/02/2020] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Given the potential of real-world evidence (RWE) to inform understanding of the risk-benefit profile of next-generation sequencing (NGS)-based testing, we undertook a study to describe the current landscape of whether and how payers use RWE as part of their coverage decision making and potential solutions for overcoming barriers. METHODS We performed a scoping literature review of existing RWE evidentiary frameworks for evaluating new technologies and identified barriers to clinical integration and evidence gaps for NGS. We synthesized findings as potential solutions for improving the relevance and utility of RWE for payer decision-making. RESULTS Payers require evidence of clinical utility to inform coverage decisions, yet we found a relatively small number of published RWE studies, and these are predominately focused on oncology, pharmacogenomics, and perinatal/pediatric testing. We identified 3 categories of innovation that may help address the current undersupply of RWE studies for NGS: (1) increasing use of RWE to inform outcomes-based contracting for new technologies, (2) precision medicine initiatives that integrate clinical and genomic data and enable data sharing, and (3) Food and Drug Administration reforms to encourage the use of RWE. Potential solutions include development of data and evidence review standards, payer engagement in RWE study design, use of incentives and partnerships to lower the barriers to RWE generation, education of payers and providers concerning the use of RWE and NGS, and frameworks for conducting outcomes-based contracting for NGS. CONCLUSIONS We provide numerous suggestions to overcome the data, methodologic, infrastructure, and policy challenges constraining greater integration of RWE in assessments of NGS.
Collapse
Affiliation(s)
| | - Michael P Douglas
- Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California at San Francisco, San Francisco, CA, USA
| | - Kathryn A Phillips
- Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California at San Francisco, San Francisco, CA, USA; Philip R. Lee Institute for Health Policy, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
22
|
Dhanda DS, Veenstra DL, Regier DA, Basu A, Carlson JJ. Payer Preferences and Willingness to Pay for Genomic Precision Medicine: A Discrete Choice Experiment. J Manag Care Spec Pharm 2020; 26:529-537. [PMID: 32223606 PMCID: PMC10390910 DOI: 10.18553/jmcp.2020.26.4.529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Although precision medicine using genetic information offers significant promise, its uptake and eventual clinical and economic impacts are uncertain. Health care payers will play an important role in evaluating evidence and costs to develop coverage and reimbursement policies. OBJECTIVE To elicit U.S. health care payer preference for genomic precision medicine to better understand trade-offs among clinical benefits, uncertainty, and cost. METHODS Using key informant interviewer discussions (N = 6 payers), we identified 6 key attributes of genetic tests important to payers: type of information the test provides (screening vs. treatment prediction), probability that the member has an informative genetic marker, expert agreement on changing medical care based on the marker, quality-of-life gains, life expectancy gains (with statistical uncertainty), and cost to the plan. We designed a stated preference discrete choice experiment using these attributes and administered a web survey to a sample of U.S. health care payers. We used effects coding and analyzed the data using an error component mixed logit modeling approach. RESULTS The survey response rate was 58% (150 participants completed the survey). Approximately 53% of respondents had previous experience evaluating genetic tests for reimbursement, and 85% had more than 5 years of health care decision-making experience. Payers valued improvements in quality of life the most (marginal willingness to pay [mWTP] of $1,513-$6,076), followed by medical expert agreement on the treatment change (mWTP of $2,881-$3,489). Payers placed a relatively lower value for genetic tests with lower marker probability (mWTP of $2,776 for highest marker probability to $423 for lowest marker probability). Payers mWTP was lowest for resolving uncertainty in quality of life (mWTP of $1,513-$2,031) and life expectancy gains ($536-$1,537). CONCLUSIONS Payers exhibited a strong preference for genetic tests that improved quality of life, had high expert agreement on changing medical care, and increased life expectancy. These findings suggest that payers will need evidence of clinical utility to support coverage and reimbursement of genomic precision medicine. DISCLOSURES This study was supported by a grant from the NIH Common Fund and NIA (1U01AG047109-01) via the Personalized Medicine Economics Research (PriMER) project. Unrelated to this study, Veenstra reports consulting fees from Bayer and Halozyme; Basu reports consulting fees from Salutis Consulting; and Reiger reports consulting fees from Roche. Carlson reports grants from Institute for Clinical and Economic Review, during the conduct of this study, and consulting fees from Bayer, Adaptive Biotechnologies, Allergan, Galderma, and Vifor Pharma, unrelated to this study.
Collapse
Affiliation(s)
- Devender S. Dhanda
- The Comparative Health Outcomes, Policy & Economics (CHOICE) Institute, University of Washington School of Pharmacy, Seattle
| | - David L. Veenstra
- The Comparative Health Outcomes, Policy & Economics (CHOICE) Institute, University of Washington School of Pharmacy, Seattle
| | - Dean A. Regier
- School of Population and Public Health, University of British Columbia, and Canadian Centre for Applied Research in Cancer Control (ARCC), BC Cancer, Vancouver, BC, Canada
| | - Anirban Basu
- The Comparative Health Outcomes, Policy & Economics (CHOICE) Institute, University of Washington School of Pharmacy, Seattle
| | - Josh J. Carlson
- The Comparative Health Outcomes, Policy & Economics (CHOICE) Institute, University of Washington School of Pharmacy, Seattle
| |
Collapse
|
23
|
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: 9.0] [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
|
24
|
Schuh MJ. Additional Utility of Pharmacogenomics (PGx) Panel Testing in a CYP2D6 Normal Metabolizer with a History of Breast Cancer. Innov Pharm 2019; 10:10.24926/iip.v10i3.2013. [PMID: 34007571 PMCID: PMC8127095 DOI: 10.24926/iip.v10i3.2013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To demonstrate the utility of pharmacogenomic (PGx) panel testing use versus single gene testing for a single indication. Panel testing may not only help further refine clinical decision making for the primary medical indication, it may uncover with one diagnostic test multiple PGx abnormalities, altering current and future therapy for other conditions. SUMMARY Breast cancer patient presented to the pharmacist PGx service to discuss results and to help determine best guidance for post-surgical pain treatment. From the panel testing it was incidentally found the patient may be at higher clot risk from standard cancer prophylactic, hormone therapy as well as possible future cardiac therapy. CONCLUSION PGx panel testing may not only uncover potential medication related problems (MRPs) with the primary medical indication being tested, it may also refine therapy for other medical problems, resulting in avoidance of future MRPs and the health care costs associated with them.
Collapse
|
25
|
Hendricks-Sturrup RM, Mazor KM, Sturm AC, Lu CY. Barriers and Facilitators to Genetic Testing for Familial Hypercholesterolemia in the United States: A Review. J Pers Med 2019; 9:jpm9030032. [PMID: 31266140 PMCID: PMC6789613 DOI: 10.3390/jpm9030032] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/22/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Familial Hypercholesterolemia (FH) is an underdiagnosed condition in the United States (US) and globally, affecting an estimated 1/250 individuals. It is a genetic risk factor for premature cardiovascular disease and is responsible for an estimated 600,000 to 1.2 million preventable vascular events. Studies show that FH genetic testing can identify a causal gene variant in 60 to 80% of clinically suspected FH cases. However, FH genetic testing is currently underutilized in clinical settings in the US despite clinical recommendations and evidence supporting its use. Reasons for underutilization are not well understood. We conducted a literature review in the PubMed/MEDLINE database and eight peer-reviewed journals. After filtering for and reviewing 2340 articles against our inclusion criteria, we included nine commentaries or expert opinions and eight empirical studies reported between January 2014 and March 2019 in our review. After applying the Consolidated Framework for Implementation Research (CFIR), we identified a total of 26 potential barriers and 15 potential facilitators (estimated barrier to facilitator ratio of 1.73). We further estimated ratios of potential barriers to facilitators for each CFIR domain (Characteristics of Intervention, Outer Setting, Inner Setting, Characteristics of Individuals, and Process). Findings derived from our systematic approach to the literature and calculations of estimated baseline ratios of barriers and facilitators can guide future research to understand FH genetic testing implementation in diverse clinical settings. Our systematic approach to the CFIR could also be used as a model to understand or compare barriers and facilitators to other evidence-based genetic testing processes in health care settings in the US and abroad.
Collapse
Affiliation(s)
- Rachele M Hendricks-Sturrup
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA 02215 USA.
| | - Kathleen M Mazor
- Meyers Primary Care Institute, Worcester, MA 01605, USA
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Amy C Sturm
- Genomic Medicine Institute, Geisinger, Danville, PA 17822, USA
| | - Christine Y Lu
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA 02215 USA
| |
Collapse
|
26
|
Hendricks-Sturrup RM, Lu CY. Understanding Implementation Challenges to Genetic Testing for Familial Hypercholesterolemia in the United States. J Pers Med 2019; 9:jpm9010009. [PMID: 30717118 PMCID: PMC6463173 DOI: 10.3390/jpm9010009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/19/2019] [Accepted: 01/21/2019] [Indexed: 01/21/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the United States (US), with familial hypercholesterolemia (FH) being a major inherited and genetic risk factor for premature CVD and atherosclerosis. Genetic testing has helped patients and providers confirm the presence of known pathogenic and likely pathogenic variations in FH-associated genes. Key organizations, such as the Centers for Disease Control and Prevention (CDC), American Heart Association (AHA), FH Foundation, and National Lipid Association (NLA), have recognized the clinical utility of FH genetic testing. However, FH genetic testing is underutilized in clinical practice in the US for reasons that are underexplored through the lens of implementation science. In this commentary, we discuss seven key implementation challenges that must be overcome to strengthen the clinical adoption of FH genetic testing in the US. These implementation challenges center on evidence of cost-effectiveness, navigating patient and provider preferences and concerns, gender and ethnic diversity and representation in genetic testing, and establishing clinical consensus around FH genetic testing based on the latest and most relevant research findings. Overcoming these implementation challenges is imperative to the mission of reducing CVD risk in the US.
Collapse
Affiliation(s)
- Rachele M Hendricks-Sturrup
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Landmark Center, 401 Park Drive Suite 401, Boston, MA 02215, USA.
| | - Christine Y Lu
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Landmark Center, 401 Park Drive Suite 401, Boston, MA 02215, USA.
| |
Collapse
|
27
|
Patterns of treatment and BRAF testing with immune checkpoint inhibitors and targeted therapy in patients with metastatic melanoma presumed to be BRAF positive. Melanoma Res 2018; 29:301-310. [PMID: 30247203 PMCID: PMC6494034 DOI: 10.1097/cmr.0000000000000504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Supplemental Digital Content is available in the text. Patients with BRAF V600 (BRAF) mutated metastatic melanoma are eligible for therapy with both immune checkpoint inhibitors and targeted therapies, making treatment choice a complex decision. The present study aimed to describe patterns of treatment with immunotherapy and targeted therapy and BRAF testing in patients with metastatic melanoma presumed to have BRAF mutations (BRAF+) in the years following the approval of the newer generation of immune checkpoint inhibitors and targeted therapies (2014–2016). Two large US commercial claims databases [Truven Health Analytics MarketScan and IQVIA Real-World Data Adjudicated Claims – USA (IQVIA RWD Adjudicated Claims – USA)] were used. Patients were presumed BRAF+ if they received at least 2 lines of therapy of which at least 1 included targeted therapy. Sequence of lines of therapy and regimens used in first (1L), second (2L), and third (3L), as well as timing of BRAF testing by sequence of therapy were described. In the Truven sample (n=162), targeted therapy was used by 66% in 1L and by 54% in 2L, and 62% had a BRAF test; in the IQVIA RWD Adjudicated Claims – USA sample (n=247), targeted therapy was used by 62% in 1L and by 50% in 2L, and 68% had a BRAF test. Among those with a claim for a BRAF test prior to 1L, over two-thirds were initiated on targeted therapy. These findings suggest that the rate of BRAF testing remained low in the years following the approval of BRAF-targeted regimens for metastatic disease. Given the recently approved adjuvant treatment options for stage III melanoma, improving the rates of BRAF testing becomes increasingly important.
Collapse
|