1
|
Haldar T, Kvale M, Yang J, Douglas MP, Coyote-Maetas W, Kachuri L, Witte JS, Iribarren C, Medina MW, Krauss RM, Yee SW, Oni-Orisan A. SLCO1B1 Functional Variants, Bilirubin, Statin-Induced Myotoxicity, and Recent Sub-Saharan African Ancestry: A Precision Medicine Health Equity Study. Clin Pharmacol Ther 2025. [PMID: 40047317 DOI: 10.1002/cpt.3624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/19/2025] [Indexed: 04/25/2025]
Abstract
Statin pharmacogenetic implementation guidelines are derived from evidence of primarily Eurocentrically biased study populations. Functional SLCO1B1 variants that are rare in these study populations have not been equitably investigated and are thus missing from guidelines. The objective of this precision medicine health equity study was to determine the clinical validity of understudied candidate functional SLCO1B1 variants common in people with 1,000 Genomes sub-Saharan African superpopulation (1KG-AFR-like) genetic similarity. We conducted our analyses using the real-world evidence of participants from three large, electronic health record-linked biobanks. We used bilirubin levels (as an endogenous substrate of organic anion transporting polypeptide [OATP1B1] function) and severe statin-induced myotoxicity phenotypes. Loss-of-function splice variant rs77271279 (P = 1.1 × 10-17) had the strongest association with elevated total bilirubin levels in Black participants (mean 84% AFR-like genetic similarity) followed by missense variant rs59502379 (P = 7.4 × 10-12) then missense variant rs4149056 (P = 6.0 × 10-5). In an exploratory subset of the Black study population who used statins (n = 77 severe statin-induced myotoxicity cases), rs59502379 (odds ratio [OR] = 2.85, 95% confidence interval [CI] 1.08-7.52), but not rs77271279 (OR = 1.75, 95% CI 0.62-4.73) was associated with myotoxicity. Sensitivity analyses in participants with >5% AFR-like genetic similarity corroborated these findings. For white participants, rs77271279 and rs59502379 were rare precluding subsequent analyses. Our findings highlight the clinical relevance for understudied SLCO1B1 variants on pharmacogenetic testing panels with a potential immediate impact on reducing the risk of severe statin-induced myotoxicity primarily in Black patients, a group historically excluded from genomic research. Future studies require larger statin user study populations with less heterogeneity by statin type.
Collapse
Affiliation(s)
- Tanushree Haldar
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, California, USA
| | - Mark Kvale
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
| | - Jia Yang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Michael P Douglas
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, California, USA
| | - Willow Coyote-Maetas
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, California, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - John S Witte
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Biomedical Data Science and Genetics, Stanford University, Stanford, California, USA
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California, USA
| | - Marisa W Medina
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
- Liver Center, University of California San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California San Francisco, Oakland, California, USA
| | - Ronald M Krauss
- Liver Center, University of California San Francisco, San Francisco, California, USA
- Department of Medicine, University of California San Francisco, Oakland, California, USA
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, California, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
2
|
Knezevic CE, Stevenson JM, Merran J, Snyder I, Restorick G, Waters C, Marzinke MA. Implementation of Integrated Clinical Pharmacogenomics Testing at an Academic Medical Center. J Appl Lab Med 2025; 10:259-273. [PMID: 39657156 DOI: 10.1093/jalm/jfae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/04/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Pharmacogenomics has demonstrated benefits for clinical care, including a reduction in adverse events and cost savings. However, barriers in expanded implementation of pharmacogenomics testing include prolonged turnaround times and integration of results into the electronic health record with clinical decision support. A clinical workflow was developed and implemented to facilitate in-house result generation and incorporation into the electronic health record at a large academic medical center. METHODS An 11-gene actionable pharmacogenomics panel was developed and validated using a QuantStudio 12K Flex platform. Allelic results were exported to a custom driver and rules engine, and result messages, which included a diplotype and predicted metabolic phenotype, were sent to the electronic health record; an electronic consultation (eConsult) service was integrated into the workflow. Postimplementation monitoring was performed to evaluate the frequency of actionable results and turnaround times. RESULTS The actionable pharmacogenomics panel covered 39 alleles across 11 genes. Metabolic phenotypes were resulted alongside gene diplotypes, and clinician-facing phenotype summaries (Genomic Indicators) were presented in the electronic health record. Postimplementation, 8 clinical areas have utilized pharmacogenomics testing, with 56% of orders occurring in the outpatient setting; 22.1% of requests included at least one actionable pharmacogene, and 67% of orders were associated with a pre- or postresult electronic consultation. Mean turnaround time from sample collection to result was 4.6 days. CONCLUSIONS A pharmacogenomics pipeline was successfully operationalized at a quaternary academic medical center, with direct integration of results into the electronic health record, clinical decision support, and eConsult services.
Collapse
Affiliation(s)
- Claire E Knezevic
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - James M Stevenson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan Merran
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Isabel Snyder
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | | | - Mark A Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Riley K, Yap K, Foley G, Lambe J, Lund S. Impact of a clinical decision support system on identifying drug-related problems and making recommendations to providers during community pharmacist-led medication reviews in Ontario, Canada: A pilot study. J Eval Clin Pract 2025; 31:e14123. [PMID: 39138861 DOI: 10.1111/jep.14123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/17/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024]
Abstract
OBJECTIVE To evaluate the impact of a clinical decision support system (CDSS) to identify drug-related problems (DRPs) during community pharmacist medication reviews. DESIGN Pilot 3-phase (group), open-label study. SETTING AND PARTICIPANTS Two community pharmacies in Sarnia, Ontario, with pharmacists providing medication reviews to patients. STUDY PROCEDURES Five pharmacists participated in three phases (groups). During Phase 1, pharmacists conducted medication reviews in 25 adult patients using the usual approaches. In Phase 2, pharmacists were trained to use a CDSS to identify DRPs, and then conducted medication reviews using the tool in a different group of 25 adult patients. In Phase 3, pharmacists conducted medication reviews without the aid of the CDSS in 25 additional adult patients. MAIN OUTCOME MEASURES The primary outcome was recommendation to the primary care physician to alter pharmacotherapy based on medication review, assessed using mean number and frequency (yes/no) of recommendations by patient. Secondary outcomes included number of potential DRPs, actual DRPs, medication review duration time, pharmacist's perceptions of the CDSS and patient satisfaction with medication review. RESULTS The mean number of recommendations to primary care physicians to alter pharmacotherapy per patient in Phases 1, 2 and 3 did not differ: 1.0 (SD = I.2) versus 1.5 (1.0) versus 1.5 (1.0), respectively; p = 0.223. The percentage of patients with a pharmacy recommendation sent to physicians across the phases, however, differed: 52% versus 80% versus 88%, respectively; p = 0.010, with more in Phases 2 and 3 compared to 1. There were more potential DRPs in group 2 compared to other groups. There were no differences in actual DRPs and medication review time. Pharmacists had positive attitudes about the CDSS. Patients were generally satisfied with their medication review. CONCLUSIONS This small pilot study provides some preliminary evidence for performance and feasibility of a CDSS to identify DRPs that pharmacists will act on. Future research is recommended to validate these findings in a larger sample.
Collapse
Affiliation(s)
- Karen Riley
- Hogan Pharmacy Partners LTD, Sarnia, Ontario, Canada
- KD Riley Pharmacist Professional Corporation, Sarnia, Ontario, Canada
| | - Katherine Yap
- Hogan Pharmacy Partners LTD, Sarnia, Ontario, Canada
| | - Gaelan Foley
- Hogan Pharmacy Partners LTD, Sarnia, Ontario, Canada
| | - John Lambe
- Hogan Pharmacy Partners LTD, Sarnia, Ontario, Canada
| | - Sean Lund
- Hogan Pharmacy Partners LTD, Sarnia, Ontario, Canada
| |
Collapse
|
4
|
Cavallari LH, Hicks JK, Patel JN, Elchynski AL, Smith DM, Bargal SA, Fleck A, Aquilante CL, Killam SR, Lemke L, Ochi T, Ramsey LB, Haidar CE, Ho T, El Rouby N, Monte AA, Allen JD, Beitelshees AL, Bishop JR, Bousman C, Campbell R, Cicali EJ, Cook KJ, Duong B, Tsermpini EE, Girdwood ST, Gregornik DB, Grimsrud KN, Lamb N, Lee JC, Lopez RO, Mazhindu TA, Morris SA, Nagy M, Nguyen J, Pasternak AL, Petry N, van Schaik RH, Schultz A, Skaar TC, Al Alshaykh H, Stevenson JM, Stone RM, Tran NK, Tuteja S, Woodahl EL, Yuan LC, Lee CR. The Pharmacogenomics Global Research Network Implementation Working Group: global collaboration to advance pharmacogenetic implementation. Pharmacogenet Genomics 2025; 35:1-11. [PMID: 39485373 PMCID: PMC11664750 DOI: 10.1097/fpc.0000000000000547] [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] [Indexed: 11/03/2024]
Abstract
Pharmacogenetics promises to optimize treatment-related outcomes by informing optimal drug selection and dosing based on an individual's genotype in conjunction with other important clinical factors. Despite significant evidence of genetic associations with drug response, pharmacogenetic testing has not been widely implemented into clinical practice. Among the barriers to broad implementation are limited guidance for how to successfully integrate testing into clinical workflows and limited data on outcomes with pharmacogenetic implementation in clinical practice. The Pharmacogenomics Global Research Network Implementation Working Group seeks to engage institutions globally that have implemented pharmacogenetic testing into clinical practice or are in the process or planning stages of implementing testing to collectively disseminate data on implementation strategies, metrics, and health-related outcomes with the use of genotype-guided drug therapy to ultimately help advance pharmacogenetic implementation. This paper describes the goals, structure, and initial projects of the group in addition to implementation priorities across sites and future collaborative opportunities.
Collapse
Affiliation(s)
- Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville
| | - J. Kevin Hicks
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Jai N. Patel
- Atrium Health Levine Cancer Institute, Charlotte
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | | | - D. Max Smith
- MedStar Health, Columbia, Maryland
- Department of Oncology, Georgetown University Medical Center, Washington, DC
| | - Salma A. Bargal
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashley Fleck
- Department of Pharmacy, Richard L. Roudebush Veterans Affairs Medical Center, Veteran Health Indiana, Indianapolis, Indiana
| | - Christina L. Aquilante
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shayna R. Killam
- L.S. Skaggs Institute for Health Innovation and Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | | | - Taichi Ochi
- Department of Pharmacotherapy, Epidemiology & Economics, Groningen Research Institute of Pharmacy; and University Library, University of Groningen, Groningen, The Netherlands
| | - Laura B. Ramsey
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, Missouri
| | - Cyrine E. Haidar
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Teresa Ho
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Nihal El Rouby
- Department of Pharmacy, St. Elizabeth HealthCare, Edgewood, Kentucky
- Division of Pharmacy Practice and Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Andrew A. Monte
- Rocky Mountain Poison & Drug Safety, Denver Health & Hospital Authority, Denver, Colorado
- University of Colorado School of Medicine, Aurora, Colorado
| | - Josiah D. Allen
- Department of Pharmacy, St. Elizabeth HealthCare, Edgewood, Kentucky
| | - Amber L. Beitelshees
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jeffrey R. Bishop
- Department of Experimental and Clinical Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chad Bousman
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
| | - Ronald Campbell
- Allegheny General Hospital, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Emily J. Cicali
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville
| | - Kelsey J. Cook
- Department of Pharmacy Education and Practice, University of Florida College of Pharmacy
- Nemours Children’s Health, Jacksonville, Florida
| | - Benjamin Duong
- Precision Medicine Program, Nemours Children’s Health Delaware Valley, Wilmington, Delaware, USA
| | - Evangelia Eirini Tsermpini
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Sonya Tang Girdwood
- Divisions of Hospital Medicine and Translational and Clinical Pharmacology, Cincinnati Children’s Hospital
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David B. Gregornik
- Pharmacogenomics Program, Children’s Minnesota, Minneapolis/St Paul, Minnesota
| | - Kristin N. Grimsrud
- Department of Pathology and Laboratory Medicine, University of California Health, Sacramento, California
| | - Nathan Lamb
- Department of Pharmacy, Ann & Robert H. Lurie Children’s Hospital of Chicago
| | - James C. Lee
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Rocio Ortiz Lopez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
| | | | - Sarah A. Morris
- Atrium Health Levine Cancer Institute, Charlotte
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Mohamed Nagy
- Personalised Medication Management Unit, Children’s Cancer Hospital Egypt 57357, Cairo, Egypt
| | - Jenny Nguyen
- Personalized Care Program, Children’s Hospital Los Angeles, Los Angeles, California
| | - Amy L. Pasternak
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan
| | - Natasha Petry
- Sanford Imagenetics, Sanford Health, Sioux Falls, South Dakota
- Department of Pharmacy Practice, North Dakota State University, Fargo, North Dakota, USA
| | - Ron H.N. van Schaik
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - April Schultz
- Sanford Imagenetics, Sanford Health, Sioux Falls, South Dakota
- Department of Internal Medicine, University of South Dakota School of Medicine, Vermillion, South Dakota
| | - Todd C. Skaar
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hana Al Alshaykh
- Pharmaceutical Care Department, King Faisal Specialist Hospital and Research Center, College of Pharmacy, Alfaisal University, Riyadh, Saudi Arabia
| | - James M. Stevenson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rachael M. Stone
- Department of Pharmacy, University of Virginia, Charlottesville, Virginia
| | - Nam K. Tran
- Department of Pathology and Laboratory Medicine, University of California Health, Sacramento, California
| | - Sony Tuteja
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Erica L. Woodahl
- L.S. Skaggs Institute for Health Innovation and Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Li-Chi Yuan
- Providence Health and Services, Irvine, California
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Mosch R, van der Lee M, Guchelaar HJ, Swen JJ. Pharmacogenetic Panel Testing: A Review of Current Practice and Potential for Clinical Implementation. Annu Rev Pharmacol Toxicol 2025; 65:91-109. [PMID: 39348848 DOI: 10.1146/annurev-pharmtox-061724-080935] [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] [Indexed: 10/02/2024]
Abstract
Pharmacogenetics (PGx) aims to optimize drug treatment outcomes by using a patient's genetic profile for individualized drug and dose selection. Currently, reactive and pretherapeutic single-gene PGx tests are increasingly applied in clinical practice in several countries and institutions. With over 95% of the population carrying at least one actionable PGx variant, and with drugs impacted by these genetic variants being in common use, pretherapeutic or preemptive PGx panel testing appears to be an attractive option for better-informed drug prescribing. Here, we discuss the current state of PGx panel testing and explore the potential for clinical implementation. We conclude that available evidence supports the implementation of pretherapeutic PGx panel testing for drugs covered in the PGx guidelines, yet identification of specific patient populations that benefit most and cost-effectiveness data are necessary to support large-scale implementation.
Collapse
Affiliation(s)
- R Mosch
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - M van der Lee
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - H J Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - J J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| |
Collapse
|
6
|
Fragala MS, Keogh M, Goldberg SE, Lorenz RA, Shaman JA. Clinical and economic outcomes of a pharmacogenomics-enriched comprehensive medication management program in a self-insured employee population. THE PHARMACOGENOMICS JOURNAL 2024; 24:30. [PMID: 39358335 PMCID: PMC11446811 DOI: 10.1038/s41397-024-00350-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 07/23/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024]
Abstract
Clinical and economic outcomes from a pharmacogenomics-enriched comprehensive medication management program were evaluated over 26 months in a self-insured U.S. employee population (n = 452 participants; n = 1500 controls) using propensity matched pre-post design with adjusted negative binomial and linear regression models. After adjusting for baseline covariates, program participation was associated with 39% fewer inpatient (p = 0.05) and 39% fewer emergency department (p = 0.002) visits, and with 21% more outpatient visits (p < 0.001) in the follow-up period compared to the control group. Results show pharmacogenomics-enriched comprehensive medication management can favorably impact healthcare utilization in a self-insured employer population by reducing emergency department and inpatient visits and can offer the potential for cost savings. Self-insured employers may consider implementing pharmacogenomics-enriched comprehensive medication management to improve the healthcare of their employees.
Collapse
|
7
|
Dong L, Zhang S, Lv C, Xue Q, Yin T. A Pharmacogenetic Panel-Based Prediction of the Clinical Outcomes in Elderly Patients with Coronary Artery Disease. Pharmaceutics 2024; 16:1079. [PMID: 39204424 PMCID: PMC11359157 DOI: 10.3390/pharmaceutics16081079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Clinical annotations for the actionable pharmacogenetic variants affecting the efficacy of cardiovascular drugs have been collected, yet their impacts on elderly patients with coronary artery disease (CAD) undergoing polypharmacy remain uncertain. We consecutively enrolled 892 elderly patients (mean age 80.7 ± 5.2) with CAD and polypharmacy. All the included patients underwent genotyping for 13 variants in 10 pharmacogenes (CYP2C19, CYP2C9, CYP4F2, CYP2D6, VKORC1, SLCO1B1, APOE, ACE, ADRB1, and MTHFR), which have the clinical annotations for 12 drugs that are commonly prescribed for patients with CAD. We found that 80.3% of the elderly CAD patients had at least one drug-gene pair associated with a therapeutical drug change. After adjusting for covariates, the number of drug-gene pairs was independently associated with a decreased risk of both major cardiovascular events (MACEs) (adjusted hazard ratio [HR]: 0.803, 95% confidence interval [CI]: 0.683-0.945, p = 0.008) and all-cause mortality (adjusted HR: 0.848, 95% CI: 0.722-0.996, p = 0.045), but also with an increased risk of adverse drug reactions (ADRs) (adjusted HR: 1.170, 95% CI: 1.030-1.329, p = 0.016). The Kaplan-Meier survival curves showed that compared to patients without a drug-gene pair, a significantly lower risk of MACEs could be observed in patients with a drug-gene pair during a 4-year follow-up (HR: 0.556, 95% CI: 0.325-0.951, p = 0.013). In conclusion, the carrier status of the actionable drug-gene pair is predictive for the clinical outcomes in elderly patients with CAD and polypharmacy. Implementing early or preemptive pharmacogenetic panel-guided polypharmacy holds the potential to enhance clinical outcomes for these patients.
Collapse
Affiliation(s)
- Lisha Dong
- Institute of Geriatrics, National Clinical Research Center for Geriatric Diseases, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (L.D.); (S.Z.); (C.L.)
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Shizhao Zhang
- Institute of Geriatrics, National Clinical Research Center for Geriatric Diseases, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (L.D.); (S.Z.); (C.L.)
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Lv
- Institute of Geriatrics, National Clinical Research Center for Geriatric Diseases, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (L.D.); (S.Z.); (C.L.)
| | - Qiao Xue
- Department of Cardiology, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Tong Yin
- Institute of Geriatrics, National Clinical Research Center for Geriatric Diseases, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (L.D.); (S.Z.); (C.L.)
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
8
|
Delabays B, Trajanoska K, Walonoski J, Mooser V. Cardiovascular Pharmacogenetics: From Discovery of Genetic Association to Clinical Adoption of Derived Test. Pharmacol Rev 2024; 76:791-827. [PMID: 39122647 DOI: 10.1124/pharmrev.123.000750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 04/24/2024] [Accepted: 05/28/2024] [Indexed: 08/12/2024] Open
Abstract
Recent breakthroughs in human genetics and in information technologies have markedly expanded our understanding at the molecular level of the response to drugs, i.e., pharmacogenetics (PGx), across therapy areas. This review is restricted to PGx for cardiovascular (CV) drugs. First, we examined the PGx information in the labels approved by regulatory agencies in Europe, Japan, and North America and related recommendations from expert panels. Out of 221 marketed CV drugs, 36 had PGx information in their labels approved by one or more agencies. The level of annotations and recommendations varied markedly between agencies and expert panels. Clopidogrel is the only CV drug with consistent PGx recommendation (i.e., "actionable"). This situation prompted us to dissect the steps from discovery of a PGx association to clinical translation. We found 101 genome-wide association studies that investigated the response to CV drugs or drug classes. These studies reported significant associations for 48 PGx traits mapping to 306 genes. Six of these 306 genes are mentioned in the corresponding PGx labels or recommendations for CV drugs. Genomic analyses also highlighted the wide between-population differences in risk allele frequencies and the individual load of actionable PGx variants. Given the high attrition rate and the long road to clinical translation, additional work is warranted to identify and validate PGx variants for more CV drugs across diverse populations and to demonstrate the utility of PGx testing. To that end, pre-emptive PGx combining genomic profiling with electronic medical records opens unprecedented opportunities to improve healthcare, for CV diseases and beyond. SIGNIFICANCE STATEMENT: Despite spectacular breakthroughs in human molecular genetics and information technologies, consistent evidence supporting PGx testing in the cardiovascular area is limited to a few drugs. Additional work is warranted to discover and validate new PGx markers and demonstrate their utility. Pre-emptive PGx combining genomic profiling with electronic medical records opens unprecedented opportunities to improve healthcare, for CV diseases and beyond.
Collapse
Affiliation(s)
- Benoît Delabays
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| | - Katerina Trajanoska
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| | - Joshua Walonoski
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| | - Vincent Mooser
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| |
Collapse
|
9
|
Sulieman L, McCoy AB, Sama L, Peterson JF. The Use of Precision Medicine to Support the Precision of Clinical Decisions in care delivery. Yearb Med Inform 2024; 33:168-174. [PMID: 40199302 PMCID: PMC12020522 DOI: 10.1055/s-0044-1800738] [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] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVES Objective: Precision medicine uses individualized patient data, including genomic and social determinants of health SDoH), to provide optimized personalized patient treatment. In this scoping review, we summarize studies published in the last two years that reported on implementation of precision medicine in clinical decision support (CDS) related to precision medicine. METHODS We searched PubMed for manuscripts published in 2022 and 2023 to retrieve publications that included CDS and precision medicine keywords and Mesh terms. We reviewed the abstracts and full texts to apply the inclusion criteria that the study must have described the implementation of precision medicine related CDS within electronic health records. We extracted the domain, type of data used in CDS, target population included in the implementation from the final set of included manuscripts. RESULTS Our search retrieved 285 manuscripts and papers. Sixteen (16) papers met inclusion criteria after manual review of the full text. Eight of the reviewed papers studied the successful implementation of pharmacogenomics in CDS, four studies investigated the implementation of disease risk, and only one paper described the implementation of CDS integrating social determinants of health. CONCLUSION Our scoping review of recent literature highlighted several findings. Pharmacogenomics is the most implemented precision medicine intervention based on published studies. Few reports describing disease risk and polygenic risk scores were found and no study addressed CDS for continuous biometric monitoring. Despite the increasing attention to social determinants of health as a key predictor of health outcomes, only one CDS incorporating SDoH have been publicly reported. Regular updates to scoping reviews can investigate barriers to implementation and identify solutions.
Collapse
Affiliation(s)
- Lina Sulieman
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Allison B. McCoy
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Lipika Sama
- Department of General Internal Medicine, Harvard Medical School, Boston, MA
| | - Josh F. Peterson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
10
|
Donnelly RS, Cavallari LH, McCune JS, Trofe-Clark J, Formea CM, Hoffecker G, Csere MM, Anderson KC, Bhat S, Mosley SA, Ma Q, Ferdock A, Hoffman JM, Hicks JK, Caudle KE. Decoding Pharmacogenomic Test Interpretation and Application to Patient Care. JOURNAL OF THE AMERICAN COLLEGE OF CLINICAL PHARMACY 2024; 7:581-588. [PMID: 39582510 PMCID: PMC11583779 DOI: 10.1002/jac5.1958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 11/26/2024]
Abstract
Pharmacogenomics is a growing area of medicine, and pharmacists across clinical practice settings have the opportunity to individualize medication selection and dosing using genetic data. However, many practicing pharmacists may feel ill-equipped to interpret pharmacogenomic test results because of insufficient education and training. Evidence-based, updated, and freely available resources such as the Clinical Pharmacogenetics Implementation Consortium guidelines can help pharmacists interpret and apply pharmacogenomic test results to patient care. Although gaps for the application of pharmacogenomic information exist, this commentary aims to demystify the interpretation of pharmacogenomic test results and empower pharmacists to apply genetic data alongside other clinical variables to optimize medication-related outcomes for their patients. An "ABCD" framework is proposed to guide pharmacists through the steps: (1) Actionability - Are the gene(s) clinically relevant for the patient? (2) Be Mindful of Limitations - What are the caveats with pharmacogenomic test results and reports? (3) Clinical Practice Guidelines - How do you use pharmacogenomic test results to guide clinical decision-making? and (4) Document and Discuss - How do you educate the patient about their pharmacogenomic test results and document the results for future use? Key concepts are illustrated using a psychiatric patient case example.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qing Ma
- American College of Clinical Pharmacy
| | | | | | | | | |
Collapse
|
11
|
Cornel MC, van der Meij KRM, van El CG, Rigter T, Henneman L. Genetic Screening-Emerging Issues. Genes (Basel) 2024; 15:581. [PMID: 38790210 PMCID: PMC11121342 DOI: 10.3390/genes15050581] [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: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
In many countries, some form of genetic screening is offered to all or part of the population, either in the form of well-organized screening programs or in a less formalized way. Screening can be offered at different phases of life, such as preconception, prenatal, neonatal and later in life. Screening should only be offered if the advantages outweigh the disadvantages. Technical innovations in testing and treatment are driving changes in the field of prenatal and neonatal screening, where many jurisdictions have organized population-based screening programs. As a result, a greater number and wider range of conditions are being added to the programs, which can benefit couples' reproductive autonomy (preconception and prenatal screening) and improve early diagnosis to prevent irreversible health damage in children (neonatal screening) and in adults (cancer and cascade screening). While many developments in screening are technology-driven, citizens may also express a demand for innovation in screening, as was the case with non-invasive prenatal testing. Relatively new emerging issues for genetic screening, especially if testing is performed using DNA sequencing, relate to organization, data storage and interpretation, benefit-harm ratio and distributive justice, information provision and follow-up, all connected to acceptability in current healthcare systems.
Collapse
Affiliation(s)
- Martina C. Cornel
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Karuna R. M. van der Meij
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Carla G. van El
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Tessel Rigter
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Lidewij Henneman
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1100 DD Amsterdam, The Netherlands
| |
Collapse
|
12
|
Shriver SP, Adams D, McKelvey BA, McCune JS, Miles D, Pratt VM, Ashcraft K, McLeod HL, Williams H, Fleury ME. Overcoming Barriers to Discovery and Implementation of Equitable Pharmacogenomic Testing in Oncology. J Clin Oncol 2024; 42:1181-1192. [PMID: 38386947 PMCID: PMC11003514 DOI: 10.1200/jco.23.01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/08/2023] [Accepted: 12/12/2023] [Indexed: 02/24/2024] Open
Abstract
Pharmacogenomics (PGx), the study of inherited genomic variation and drug response or safety, is a vital tool in precision medicine. In oncology, testing to identify PGx variants offers patients the opportunity for customized treatments that can minimize adverse effects and maximize the therapeutic benefits of drugs used for cancer treatment and supportive care. Because individuals of shared ancestry share specific genetic variants, PGx factors may contribute to outcome disparities across racial and ethnic categories when genetic ancestry is not taken into account or mischaracterized in PGx research, discovery, and application. Here, we examine how the current scientific understanding of the role of PGx in differential oncology safety and outcomes may be biased toward a greater understanding and more complete clinical implementation of PGx for individuals of European descent compared with other genetic ancestry groups. We discuss the implications of this bias for PGx discovery, access to care, drug labeling, and patient and provider understanding and use of PGx approaches. Testing for somatic genetic variants is now the standard of care in treatment of many solid tumors, but the integration of PGx into oncology care is still lacking despite demonstrated actionable findings from PGx testing, reduction in avoidable toxicity and death, and return on investment from testing. As the field of oncology is poised to expand and integrate germline genetic variant testing, it is vital that PGx discovery and application are equitable for all populations. Recommendations are introduced to address barriers to facilitate effective and equitable PGx application in cancer care.
Collapse
Affiliation(s)
| | | | | | - Jeannine S McCune
- City of Hope/Beckman Research Institute Department of Hematologic Malignancies Translational Sciences, Duarte, CA
| | | | | | | | | | | | | |
Collapse
|
13
|
Abouelhoda M, Almuqati N, Abogosh A, Alfraih F, Maddirevula S, Alkuraya FS. Mining local exome and HLA data to characterize pharmacogenetic variants in Saudi Arabia. Hum Genet 2024; 143:125-136. [PMID: 38159139 DOI: 10.1007/s00439-023-02628-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
Pharmacogenomics (PGx) is a promising field of precision medicine where efficacy of drugs is maximized while side effects are minimized for individual patients. Knowledge of the frequency of PGx-relevant variants (pharmacovariants) in the local population is a pre-requisite to informed policy making. Unfortunately, such knowledge is largely lacking from the Middle East. Here, we describe the use of a large clinical exome database (n = 13,473) and HLA haplotypes (n = 64,737) from Saudi Arabia, one of the largest countries in the Middle East, along with previously published data from the local population to ascertain allele frequencies of known pharmacovariants. In addition, we queried another exome database (n = 816) of well-phenotyped research subjects from Saudi Arabia to discover novel candidate variants in known PGx genes (pharmacogenes). Although our results show that only 26% (63/242) of class 1A/1B PharmGKB variants were identified, we estimate that 99.57% of the local population have at least one such variant. This translates to a minimum estimated impact of 9% of medications dispensed by our medical center annually. We also highlight the contribution of rare variants where 71% of the pharmacogenes devoid of common pharmacovariants had at least one potentially deleterious rare variant. Thus, we show that approaches that go beyond the use of commercial PGx kits that have been optimized for other populations should be implemented to ensure universal and equitable access of all members of the local population to personalized prescription practices.
Collapse
Affiliation(s)
- Mohamed Abouelhoda
- Department of Computational Sciences, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Noura Almuqati
- Department of Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ahmed Abogosh
- Department of Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Feras Alfraih
- Oncology Centre, Faisal Specialist Hospital and Research Centre, Riyadh, King, Saudi Arabia
| | - Sateesh Maddirevula
- Department of Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
- Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia.
| |
Collapse
|
14
|
Tremmel R, Hofmann U, Haag M, Schaeffeler E, Schwab M. Circulating Biomarkers Instead of Genotyping to Establish Metabolizer Phenotypes. Annu Rev Pharmacol Toxicol 2024; 64:65-87. [PMID: 37585662 DOI: 10.1146/annurev-pharmtox-032023-121106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Pharmacogenomics (PGx) enables personalized treatment for the prediction of drug response and to avoid adverse drug reactions. Currently, PGx mainly relies on the genetic information of absorption, distribution, metabolism, and excretion (ADME) targets such as drug-metabolizing enzymes or transporters to predict differences in the patient's phenotype. However, there is evidence that the phenotype-genotype concordance is limited. Thus, we discuss different phenotyping strategies using exogenous xenobiotics (e.g., drug cocktails) or endogenous compounds for phenotype prediction. In particular, minimally invasive approaches focusing on liquid biopsies offer great potential to preemptively determine metabolic and transport capacities. Early studies indicate that ADME phenotyping using exosomes released from the liver is reliable. In addition, pharmacometric modeling and artificial intelligence improve phenotype prediction. However, further prospective studies are needed to demonstrate the clinical utility of individualized treatment based on phenotyping strategies, not only relying on genetics. The present review summarizes current knowledge and limitations.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany
- Departments of Clinical Pharmacology, and Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Heidelberg (DKFZ), Partner Site, Tübingen, Germany
| |
Collapse
|
15
|
Yee SW, Haldar T, Kvale M, Yang J, Douglas MP, Oni-Orisan A. SLCO1B1 functional variants and statin-induced myopathy in people with recent genealogical ancestors from Africa: a population-based real-world study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.02.23299324. [PMID: 38076949 PMCID: PMC10705643 DOI: 10.1101/2023.12.02.23299324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Background Clinical pharmacogenetic implementation guidelines for statin therapy are derived from evidence of primarily Eurocentric study populations. Functional SLCO1B1 variants that are rare in these study populations have not been investigated as a determinant of statin myotoxicity and are thus missing from guideline inclusion. Objective Determine the relationship between candidate functional SLCO1B1 variants and statin-induced myopathy in people with recent genealogical ancestors from Africa. Design Population-based pharmacogenetic study using real-world evidence from electronic health record-linked biobanks. Setting Various health care settings. Participants Self-identified white and Black statin users with genome-wide genotyping data available. Measurements Primarily, the odds of statin-induced myopathy + rhabdomyolysis. Secondarily, total bilirubin levels. Thirdly, cell-based functional assay results. Results Meta-analyses results demonstrated an increased risk of statin-induced myopathy + rhabdomyolysis with c.481+1G>T (odds ratio [OR] = 3.27, 95% confidence interval [CI] 1.43-7.46, P =.005) and c.1463G>C (OR = 2.45, 95% CI 1.04-5.78, P =.04) for Black participants. For White participants, c.521T>C was also significantly associated with increased risk of statin-induced myopathy + rhabdomyolysis (OR = 1.41, 95% CI 1.20-1.67, P =5.4x10 -5 ). This effect size for c.521T>C was similar in the Black participants, but did not meet the level of statistical significance (OR = 1.47, 95% CI 0.58-3.73, P =0.41). Supporting evidence using total bilirubin as an endogenous biomarker of SLCO1B1 function as well as from cell-based functional studies corroborated these findings. Limitations Data limited to severe statin myotoxicity events. Conclusion Our findings implicate Afrocentric SLCO1B1 variants on preemptive pharmacogenetic testing panels, which could have an instant impact on reducing the risk of statin-associated myotoxicity in historically excluded groups. Primary Funding Source National Institutes of Health, Office of the Director - All of Us (OD-AoURP).
Collapse
|
16
|
Sainz de Medrano Sainz JI, Brunet Serra M. Influencia de la farmacogenética en la diversidad de respuesta a las estatinas asociada a las reacciones adversas. ADVANCES IN LABORATORY MEDICINE 2023; 4:353-364. [PMID: 38106494 PMCID: PMC10724860 DOI: 10.1515/almed-2023-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/15/2023] [Indexed: 12/19/2023]
Abstract
Introducción Las estatinas son unos de los medicamentos más prescritos en los países desarrollados por ser el tratamiento de elección para reducir los niveles de colesterol ayudando así a prevenir la enfermedad cardiovascular. Sin embargo, un gran número de pacientes sufre reacciones adversas, en especial miotoxicidad. Entre los factores que influyen en la diversidad de respuesta, la farmacogenética puede jugar un papel relevante especialmente en la prevención de los efectos adversos asociados a estos medicamentos. Contenido Revisión de los conocimientos actuales sobre la influencia de la farmacogenética en la aparición y prevención de las reacciones adversas asociadas a estatinas, así como del beneficio clínico del test farmacogenético anticipado. Resumen Variaciones genéticas en SLCO1B1 (rs4149056) para todas las estatinas; en ABCG2 (rs2231142) para rosuvastatina; o en CYP2C9 (rs1799853 y rs1057910) para fluvastatina están asociadas a un incremento de las reacciones adversas de tipo muscular y a una baja adherencia al tratamiento. Además, diversos fármacos inhibidores de estos transportadores y enzimas de biotransformación incrementan la exposición sistémica de las estatinas favoreciendo la aparición de las reacciones adversas. Perspectiva La implementación clínica del análisis anticipado de este panel de farmacogenética evitaría en gran parte la aparición de reacciones adversas. Además, la estandarización en la identificación de los efectos adversos, en la metodología e interpretación del genotipo, permitirá obtener resultados más concluyentes sobre la asociación entre las variantes genéticas del SLCO1B1, ABCG y CYP2C9 y la aparición de reacciones adversas y establecer recomendaciones para alcanzar tratamientos más personalizados para cada estatina.
Collapse
Affiliation(s)
- Jaime I. Sainz de Medrano Sainz
- Servicio de Bioquímica y Genética Molecular, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, España
| | - Mercè Brunet Serra
- Jefa de sección de Farmacología y Toxicología, Servicio de Bioquímica y Genética Molecular, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, España
| |
Collapse
|
17
|
Sainz de Medrano Sainz JI, Brunet Serra M. Influence of pharmacogenetics on the diversity of response to statins associated with adverse drug reactions. ADVANCES IN LABORATORY MEDICINE 2023; 4:341-352. [PMID: 38106499 PMCID: PMC10724874 DOI: 10.1515/almed-2023-0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/15/2023] [Indexed: 12/19/2023]
Abstract
Background Statins are one of the most prescribed medications in developed countries as the treatment of choice for reducing cholesterol and preventing cardiovascular diseases. However, a large proportion of patients experience adverse drug reactions, especially myotoxicity. Among the factors that influence the diversity of response, pharmacogenetics emerges as a relevant factor of influence in inter-individual differences in response to statins and can be useful in the prevention of adverse drug effects. Content A systematic review was performed of current knowledge of the influence of pharmacogenetics on the occurrence and prevention of statin-associated adverse reactions and clinical benefits of preemptive pharmacogenetics testing. Summary Genetic variants SLCO1B1 (rs4149056) for all statins; ABCG2 (rs2231142) for rosuvastatin; or CYP2C9 (rs1799853 and rs1057910) for fluvastatin are associated with an increase in muscle-related adverse effects and poor treatment adherence. Besides, various inhibitors of these transporters and biotransformation enzymes increase the systemic exposure of statins, thereby favoring the occurrence of adverse drug reactions. Outlook The clinical preemptive testing of this pharmacogenetic panel would largely prevent the incidence of adverse drug reactions. Standardized methods should be used for the identification of adverse effects and the performance and interpretation of genotyping test results. Standardization would allow to obtain more conclusive results about the association between SLCO1B1, ABCG and CYP2C9 variants and the occurrence of adverse drug reactions. As a result, more personalized recommendations could be established for each statin.
Collapse
Affiliation(s)
- Jaime I. Sainz de Medrano Sainz
- Servicio de Bioquímica y Genética Molecular, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Mercè Brunet Serra
- Jefa de sección de Farmacología y Toxicología, Servicio de Bioquímica y Genética Molecular, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Van Heukelom J, Morgan J, Massmann A, Jacobsen K, Petry NJ, Baye JF, Frear S, Schultz A. Evolution of pharmacogenomic services and implementation of a multi-state pharmacogenomics clinic across a large rural healthcare system. Front Pharmacol 2023; 14:1274165. [PMID: 38035031 PMCID: PMC10682150 DOI: 10.3389/fphar.2023.1274165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction: Pharmacogenomics (PGx) aims to maximize drug benefits while minimizing risk of toxicity. Although PGx has proven beneficial in many settings, clinical uptake lags. Lack of clinician confidence and limited availability of PGx testing can deter patients from completing PGx testing. A few novel PGx clinic models have been described as a way to incorporate PGx testing into the standard of care. Background: A PGx clinic was implemented to fill an identified gap in provider availability, confidence, and utilization of PGx across our health system. Through a joint pharmacist and Advanced Practice Provider (APP) collaborative clinic, patients received counseling and PGx medication recommendations both before and after PGx testing. The clinic serves patients both in-person and virtually across four states in the upper Midwest. Results: The majority of patients seen in the PGx clinic during the early months were clinician referred (77%, n = 102) with the remainder being self-referred. Patients were, on average, taking two medications with Clinical Pharmacogenetics Implementation Consortium guidelines. Visits were split almost equally between in-person and virtual visits. Conclusion: Herein, we describe the successful implementation of an interdisciplinary PGx clinic to further enhance our PGx program. Throughout the implementation of the PGx clinic we have learned valuable lessons that may be of interest to other implementors. Clinicians were actively engaged in clinic referrals and early adoption of telemedicine was key to the clinic's early successes.
Collapse
Affiliation(s)
- Joel Van Heukelom
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
- Department of Internal Medicine, University of South Dakota School of Medicine, Vermillion, SD, United States
| | - Jennifer Morgan
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
- Department of Medical Genetics, Sanford Health, Sioux Falls, SD, United States
| | - Amanda Massmann
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
- Department of Internal Medicine, University of South Dakota School of Medicine, Vermillion, SD, United States
| | - Kristen Jacobsen
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
| | - Natasha J. Petry
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
- Department of Pharmacy Practice, North Dakota State University, Fargo, ND, United States
| | - Jordan F. Baye
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
- Department of Internal Medicine, University of South Dakota School of Medicine, Vermillion, SD, United States
- Department of Pharmacy Practice, South Dakota State University College of Pharmacy and Allied Health Professions, Brookings, SD, United States
| | - Samantha Frear
- Translational Software, Inc., Mercer Island, WA, United States
| | - April Schultz
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, United States
- Department of Internal Medicine, University of South Dakota School of Medicine, Vermillion, SD, United States
| |
Collapse
|
19
|
Rajkovic A. Pharmacogenomics and Personalized Medicine for Neonatal Care. J Pediatr 2023; 261:113575. [PMID: 37353149 DOI: 10.1016/j.jpeds.2023.113575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Affiliation(s)
- Aleksandar Rajkovic
- Department of Pathology and the Institute of Human Genetics University of California San Francisco, San Francisco, CA
| |
Collapse
|
20
|
Murry LT, Hillman LA, Allen JD, Bishop JR. Intersection and Considerations for Patient-Centered Care, Patient Experience, and Medication Experience in Pharmacogenomics. PHARMACY 2023; 11:146. [PMID: 37736918 PMCID: PMC10514786 DOI: 10.3390/pharmacy11050146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
As healthcare continues to embrace the concept of person- and patient-centered care, pharmacogenomics, patient experience, and medication experience will continue to play an increasingly important role in care delivery. This review highlights the intersection between these concepts and provides considerations for patient-centered medication and pharmacogenomic experiences. Elements at the patient, provider, and system level can be considered in the discussion, supporting the use of pharmacogenomics, with components of the patient and medication experience contributing to the mitigation of barriers surrounding patient use and the valuation of pharmacogenomic testing.
Collapse
Affiliation(s)
- Logan T. Murry
- College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA
| | - Lisa A. Hillman
- College of Pharmacy, The University of Minnesota, Minneapolis, MN 55455, USA; (L.A.H.); or (J.D.A.); (J.R.B.)
| | - Josiah D. Allen
- College of Pharmacy, The University of Minnesota, Minneapolis, MN 55455, USA; (L.A.H.); or (J.D.A.); (J.R.B.)
- Department of Pharmacy, St. Elizabeth Healthcare, Edgewood, KY 41017, USA
| | - Jeffrey R. Bishop
- College of Pharmacy, The University of Minnesota, Minneapolis, MN 55455, USA; (L.A.H.); or (J.D.A.); (J.R.B.)
- Medical School, The University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
Norris M, Dalton R, Alam B, Eddy E, Nguyen KA, Cavallari LH, Sumfest J, Wiisanen K, Cicali EJ. Lessons from clinical implementation of a preemptive pharmacogenetic panel as part of a testing pilot program with an employer-sponsored medical plan. Front Genet 2023; 14:1249003. [PMID: 37680199 PMCID: PMC10482099 DOI: 10.3389/fgene.2023.1249003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction: This manuscript reports on a pilot program focused on implementing pharmacogenetic testing within the framework of an employer-sponsored medical plan at University of Florida (UF) Health. The aim was to understand the challenges associated with program implementation and to gather insights into patient attitudes towards PGx testing. Methods: The pilot program adopted a partially preemptive approach, targeting patients on current prescriptions for medications with relevant gene-drug associations. Patients were contacted via phone or through the MyChart system and offered pharmacogenetic testing with no additional direct costs. Results: Of 244 eligible patients, 110 agreed to participate. However, only 61 returned the mailed DNA collection kits. Among these, 89% had at least one potentially actionable genotype-based phenotype. Post-test follow-up revealed that while the majority viewed the process positively, 71% preferred a consultation with a pharmacogenetic specialist for better understanding of their results. Barriers to implementation ranged from fatigue with the healthcare system to a lack of understanding of the pharmacogenetic testing and concerns about privacy and potential misuse of genetic data. Conclusion: The findings underscore the need for clearer patient education on pharmacogenetic results and suggest the importance of the role of pharmacogenetic-trained pharmacists in delivering this education. They also highlight issues with relying on incomplete or inaccurate medication lists in patients' electronic health record. The implementation revealed less obvious challenges, the understanding of which could be beneficial for the success of future preemptive pharmacogenetic implementation programs. The insights from the pilot program served to bridge the information gap between patients, providers, and pharmacogenetic -specialists, with the ultimate goal of improving patient care.
Collapse
Affiliation(s)
- Madeline Norris
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Rachel Dalton
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Benish Alam
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Elizabeth Eddy
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Khoa A. Nguyen
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Jill Sumfest
- GatorCare Health Management Corporation, University of Florida Health, Gainesville, FL, United States
| | - Kristin Wiisanen
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Emily J. Cicali
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States
- Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
22
|
Rigobello R, Shaw J, Ilg D, Zimmerman R, Edelmann L, Kornreich R, Scott SA, Cody N. Clinical Pharmacogenomic MT-RNR1 Screening for Aminoglycoside-Induced Ototoxicity and the Post-Test Counseling Conundrum. Clin Pharmacol Ther 2023; 114:262-265. [PMID: 37314952 DOI: 10.1002/cpt.2910] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/14/2023] [Indexed: 06/16/2023]
Abstract
Aminoglycoside antibiotic exposure can result in ototoxicity and irreversible hearing loss among individuals that harbor the m.1555A>G variant in the mitochondrial 12S rRNA gene, MT-RNR1. Importantly, pre-emptive m.1555A>G screening has been shown to reduce the prevalence of pediatric aminoglycoside-induced ototoxicity; however, professional guidelines to support and guide post-test pharmacogenomic counseling in this context are not currently available. This Perspective highlights key issues with delivering MT-RNR1 results, including longitudinal familial care considerations and communicating m.1555A>G heteroplasmy.
Collapse
Affiliation(s)
| | - Jay Shaw
- GeneDx, Gaithersburg, Maryland, USA
| | | | | | - Lisa Edelmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Stuart A Scott
- Department of Pathology, Stanford University, Stanford, California, USA
- Clinical Genomics Laboratory, Stanford Medicine, Palo Alta, California, USA
| | - Neal Cody
- GeneDx, Gaithersburg, Maryland, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
23
|
Agulló L, Aguado I, Muriel J, Margarit C, Gómez A, Escorial M, Sánchez A, Fernández A, Peiró AM. Pharmacogenetic Guided Opioid Therapy Improves Chronic Pain Outcomes and Comorbid Mental Health: A Randomized, Double-Blind, Controlled Study. Int J Mol Sci 2023; 24:10754. [PMID: 37445931 DOI: 10.3390/ijms241310754] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Interindividual variability in analgesic response is at least partly due to well-characterized polymorphisms that are associated with opioid dosing and adverse outcomes. The Clinical Pharmacogenetics Implementation Consortium (CPIC) has put forward recommendations for the CYP2D6 phenotype, but the list of studied drug-gene pairs continues to grow. This clinical trial randomized chronic pain patients (n = 60), referred from primary care to pain unit care into two opioid prescribing arms, one guided by CYP2D6, μ-opioid receptor (OPRM1), and catechol-O-methyl transferase (COMT) genotypes vs. one with clinical routine. The genotype-guided treatment reduced pain intensity (76 vs. 59 mm, p < 0.01) by improving pain relief (28 vs. 48 mm, p < 0.05), increased quality of life (43 vs. 56 mm p < 0.001), and lowered the incidence of clinically relevant adverse events (3 [1-5] vs. 1 [0-2], p < 0.01) and 42% opioid dose (35 [22-61] vs. 60 [40-80] mg/day, p < 0.05) as opposed to usual prescribing arm. The final health utility score was significantly higher (0.71 [0.58-0.82] vs. 0.51 [0.13-0.67] controls, p < 0.05) by improving sleepiness and depression comorbidity, with a significant reduction of 30-34% for headache, dry mouth, nervousness, and constipation. A large-scale implementation analysis could help clinical translation, together with a pharmaco-economic evaluation.
Collapse
Affiliation(s)
- Laura Agulló
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Isidro Aguado
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
| | - Javier Muriel
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - César Margarit
- Pain Unit, Department of Health of Alicante, General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
| | - Alba Gómez
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Mónica Escorial
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Astrid Sánchez
- San Vicente del Raspeig II Health Center, c/Alicante, 78, Sant Vicent del Raspeig, 03690 Alicante, Spain
| | - Alicia Fernández
- San Vicente del Raspeig II Health Center, c/Alicante, 78, Sant Vicent del Raspeig, 03690 Alicante, Spain
| | - Ana M Peiró
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| |
Collapse
|
24
|
Katsukunya JN, Soko ND, Naidoo J, Rayner B, Blom D, Sinxadi P, Chimusa ER, Dandara M, Dzobo K, Jones E, Dandara C. Pharmacogenomics of Hypertension in Africa: Paving the Way for a Pharmacogenetic-Based Approach for the Treatment of Hypertension in Africans. Int J Hypertens 2023; 2023:9919677. [PMID: 38633331 PMCID: PMC11022520 DOI: 10.1155/2023/9919677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 04/19/2024] Open
Abstract
In Africa, the burden of hypertension has been rising at an alarming rate for the last two decades and is a major cause for cardiovascular disease (CVD) mortality and morbidity. Hypertension is characterised by elevated blood pressure (BP) ≥ 140/90 mmHg. Current hypertension guidelines recommend the use of antihypertensives belonging to the following classes: calcium channel blockers (CCB), angiotensin converting inhibitors (ACEI), angiotensin receptor blockers (ARB), diuretics, β-blockers, and mineralocorticoid receptor antagonists (MRAs), to manage hypertension. Still, a considerable number of hypertensives in Africa have their BP uncontrolled due to poor drug response and remain at the risk of CVD events. Genetic factors are a major contributing factor, accounting for 20% to 80% of individual variability in therapy and poor response. Poor response to antihypertensive drug therapy is characterised by elevated BPs and occurrence of adverse drug reactions (ADRs). As a result, there have been numerous studies which have examined the role of genetic variation and its influence on antihypertensive drug response. These studies are predominantly carried out in non-African populations, including Europeans and Asians, with few or no Africans participating. It is important to note that the greatest genetic diversity is observed in African populations as well as the highest prevalence of hypertension. As a result, this warrants a need to focus on how genetic variation affects response to therapeutic interventions used to manage hypertension in African populations. In this paper, we discuss the implications of genetic diversity in CYP11B2, GRK4, NEDD4L, NPPA, SCNN1B, UMOD, CYP411, WNK, CYP3A4/5, ACE, ADBR1/2, GNB3, NOS3, B2, BEST3, SLC25A31, LRRC15 genes, and chromosome 12q loci on hypertension susceptibility and response to antihypertensive therapy. We show that African populations are poorly explored genetically, and for the few characterised genes, they exhibit qualitative and quantitative differences in the profile of pharmacogene variants when compared to other ethnic groups. We conclude by proposing prioritization of pharmacogenetics research in Africa and possible adoption of pharmacogenetic-guided therapies for hypertension in African patients. Finally, we outline the implications, challenges, and opportunities these studies present for populations of non-European descent.
Collapse
Affiliation(s)
- Jonathan N. Katsukunya
- Division of Human Genetics, Department of Pathology and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| | - Nyarai D. Soko
- Division of Human Genetics, Department of Pathology and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| | - Jashira Naidoo
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Brian Rayner
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dirk Blom
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Lipidology and Cape Heart Institute, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Phumla Sinxadi
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Clinical Pharmacology, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, Tyne and Wear NE1 8ST, UK
| | - Michelle Dandara
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| | - Kevin Dzobo
- Medical Research Council-SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences University of Cape Town, Anzio Road Observatory, Cape Town 7925, South Africa
| | - Erika Jones
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
25
|
Chambal M, Forsthoffer C, Egnaczyk A, Seitz M, Grande K, Ashcraft K, Wick JA, Blaxall BC. Comparison of targeted vs. expanded pharmacogenomic testing: What are we missing? J Am Pharm Assoc (2003) 2023; 63:939-945. [PMID: 37024375 DOI: 10.1016/j.japh.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND Pharmacogenomics (PGx) is used as a medication management strategy by a small but growing number of institutions. PGx allows prescribers to individually treat patients concordant with their genes. Recent litigation for preventable PGx-mediated adverse events highlights the need to accelerate PGx implementation for patient safety. Genetic variations cause drug metabolism, transport, and target changes, affecting medication response and tolerability. PGx testing often consists of targeted testing aimed at specific gene-drug pairs or disease states. Conversely, expanded panel testing can evaluate all known actionable gene-drug interactions, enhancing proactive clarity regarding patient response. OBJECTIVES Evaluate the divergence of targeted PGx testing with a single gene-drug pair test (cardiac), a two-gene panel, and a focused psychiatric panel compared to expanded PGx testing. METHODS An expanded PGx panel (≥25 genes) was compared to a single gene-drug pair test of CYP2C19/clopidogrel, a dual gene test of CYP2C19/CYP2D6, a 7-gene psychiatric list, and a 14-gene psychiatric panel to inform specific depression and pain management drugs. The expanded panel provided a baseline to evaluate total PGx variations compared to those possibly missed by targeted testing. RESULTS Targeted testing did not identify up to 95% of total PGx gene-drug interactions discovered. The expanded panel reported all gene-drug interactions for any medication with Clinical Pharmacogenomics Implementation Consortium (CPIC) guidance or U.S. Food and Drug Administration (FDA) labeling for that gene. Single gene CYP2C19/clopidogrel testing missed or did not report on ∼95% of total interactions, CYP2C19/CYP2D6 testing missed or did not report ∼89%, and the 14-gene panel missed or did not report on ∼73%. The 7-gene list missed ∼20% of discovered potential PGx interactions but was not designed to identify gene-drug interactions. CONCLUSIONS Targeted PGx testing for limited genes or by specialty may miss or not report significant portions of PGx gene-drug interactions. This can lead to potential patient harm from the missed interactions and subsequent failed therapies and/or adverse reactions.
Collapse
|
26
|
Jessop JP, Russell J, DeJesus A, Bardolia C, Hanna A, Turgeon J, Michaud V, Amin NS. Pharmacogenetic Testing in a 70-Year-Old Woman with Polypharmacy and Multiple Comorbidities: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2023; 24:e938850. [PMID: 36804920 PMCID: PMC9969360 DOI: 10.12659/ajcr.938850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Comorbidities and polypharmacy are difficult to manage, as polypharmacy hinders identification and prevention of medication-related problems. Risk for adverse drug events (ADEs) can be minimized through pharmacogenomic (PGx) testing and related therapeutic adjustments. CASE REPORT A 70-year-old woman with comorbidities and medications enrolled in the Program of All-inclusive Care for the Elderly presented with left lower extremity (LLE) pain, generalized weakness, and major depressive disorder. The provider requested a medication safety review, where the clinical pharmacist-recommended PGx testing given the LLE pain and weakness while taking a statin and inconsistent INR readings taking warfarin. The pharmacist recommended switching atorvastatin to pravastatin to minimize the risk for statin-associated ADEs due to CYP3A4 inhibition and switching fluoxetine to citalopram due to uncontrolled depression/anxiety and to mitigate drug-drug interactions with carvedilol to reduce the risk of orthostatic hypotension. Recommendations were accepted and upon follow-up the patient reported minor LLE pain and improved wellbeing on citalopram. Following PGx testing, the patient had decreased function at SLCO1B1 and was an intermediate metabolizer for CYP2C9 and CYP2D6. This case demonstrates how preemptive PGx testing would have identified drug-gene interactions (DGIs) at the time of prescribing and reduced the risk of statin-associated muscular symptoms, highlighting the utility of panel-based PGx testing in older adults at high risk for ADEs and/or therapy failure. CONCLUSIONS Decreased function at SLCO1B1 increases exposure to statins, leading to statin-induced myalgias, as displayed in this case. PGx testing can help identify DGIs, choose optimal therapies in medically complex older adults, and minimize ADE risk.
Collapse
Affiliation(s)
- Jayson P. Jessop
- Office of Translational Research and Residency Programs, Tabula Rasa HealthCare, Moorestown, NJ, USA
| | - Joshua Russell
- Office of Translational Research and Residency Programs, Tabula Rasa HealthCare, Moorestown, NJ, USA
| | - Adriana DeJesus
- Office of Translational Research and Residency Programs, Tabula Rasa HealthCare, Moorestown, NJ, USA
| | - Chandni Bardolia
- Office of Translational Research and Residency Programs, Tabula Rasa HealthCare, Moorestown, NJ, USA
| | - Abeer Hanna
- Program of All-Inclusive Care for the Elderly (PACE), VieCare Beaver, Pittsburgh, PA, USA
| | - Jacques Turgeon
- Precision Pharmacotherapy Research and Development Institute, Tabula Rasa HealthCare, Orlando, FL, USA,Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada,Corresponding Author: Jacques Turgeon, e-mail:
| | - Veronique Michaud
- Precision Pharmacotherapy Research and Development Institute, Tabula Rasa HealthCare, Orlando, FL, USA,Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Nishita S. Amin
- Office of Translational Research and Residency Programs, Tabula Rasa HealthCare, Moorestown, NJ, USA
| |
Collapse
|
27
|
Swen JJ, van der Wouden CH, Manson LE, Abdullah-Koolmees H, Blagec K, Blagus T, Böhringer S, Cambon-Thomsen A, Cecchin E, Cheung KC, Deneer VH, Dupui M, Ingelman-Sundberg M, Jonsson S, Joefield-Roka C, Just KS, Karlsson MO, Konta L, Koopmann R, Kriek M, Lehr T, Mitropoulou C, Rial-Sebbag E, Rollinson V, Roncato R, Samwald M, Schaeffeler E, Skokou M, Schwab M, Steinberger D, Stingl JC, Tremmel R, Turner RM, van Rhenen MH, Dávila Fajardo CL, Dolžan V, Patrinos GP, Pirmohamed M, Sunder-Plassmann G, Toffoli G, Guchelaar HJ. A 12-gene pharmacogenetic panel to prevent adverse drug reactions: an open-label, multicentre, controlled, cluster-randomised crossover implementation study. Lancet 2023; 401:347-356. [PMID: 36739136 DOI: 10.1016/s0140-6736(22)01841-4] [Citation(s) in RCA: 233] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND The benefit of pharmacogenetic testing before starting drug therapy has been well documented for several single gene-drug combinations. However, the clinical utility of a pre-emptive genotyping strategy using a pharmacogenetic panel has not been rigorously assessed. METHODS We conducted an open-label, multicentre, controlled, cluster-randomised, crossover implementation study of a 12-gene pharmacogenetic panel in 18 hospitals, nine community health centres, and 28 community pharmacies in seven European countries (Austria, Greece, Italy, the Netherlands, Slovenia, Spain, and the UK). Patients aged 18 years or older receiving a first prescription for a drug clinically recommended in the guidelines of the Dutch Pharmacogenetics Working Group (ie, the index drug) as part of routine care were eligible for inclusion. Exclusion criteria included previous genetic testing for a gene relevant to the index drug, a planned duration of treatment of less than 7 consecutive days, and severe renal or liver insufficiency. All patients gave written informed consent before taking part in the study. Participants were genotyped for 50 germline variants in 12 genes, and those with an actionable variant (ie, a drug-gene interaction test result for which the Dutch Pharmacogenetics Working Group [DPWG] recommended a change to standard-of-care drug treatment) were treated according to DPWG recommendations. Patients in the control group received standard treatment. To prepare clinicians for pre-emptive pharmacogenetic testing, local teams were educated during a site-initiation visit and online educational material was made available. The primary outcome was the occurrence of clinically relevant adverse drug reactions within the 12-week follow-up period. Analyses were irrespective of patient adherence to the DPWG guidelines. The primary analysis was done using a gatekeeping analysis, in which outcomes in people with an actionable drug-gene interaction in the study group versus the control group were compared, and only if the difference was statistically significant was an analysis done that included all of the patients in the study. Outcomes were compared between the study and control groups, both for patients with an actionable drug-gene interaction test result (ie, a result for which the DPWG recommended a change to standard-of-care drug treatment) and for all patients who received at least one dose of index drug. The safety analysis included all participants who received at least one dose of a study drug. This study is registered with ClinicalTrials.gov, NCT03093818 and is closed to new participants. FINDINGS Between March 7, 2017, and June 30, 2020, 41 696 patients were assessed for eligibility and 6944 (51·4 % female, 48·6% male; 97·7% self-reported European, Mediterranean, or Middle Eastern ethnicity) were enrolled and assigned to receive genotype-guided drug treatment (n=3342) or standard care (n=3602). 99 patients (52 [1·6%] of the study group and 47 [1·3%] of the control group) withdrew consent after group assignment. 652 participants (367 [11·0%] in the study group and 285 [7·9%] in the control group) were lost to follow-up. In patients with an actionable test result for the index drug (n=1558), a clinically relevant adverse drug reaction occurred in 152 (21·0%) of 725 patients in the study group and 231 (27·7%) of 833 patients in the control group (odds ratio [OR] 0·70 [95% CI 0·54-0·91]; p=0·0075), whereas for all patients, the incidence was 628 (21·5%) of 2923 patients in the study group and 934 (28·6%) of 3270 patients in the control group (OR 0·70 [95% CI 0·61-0·79]; p <0·0001). INTERPRETATION Genotype-guided treatment using a 12-gene pharmacogenetic panel significantly reduced the incidence of clinically relevant adverse drug reactions and was feasible across diverse European health-care system organisations and settings. Large-scale implementation could help to make drug therapy increasingly safe. FUNDING European Union Horizon 2020.
Collapse
Affiliation(s)
- Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, Netherlands
| | | | - Lisanne En Manson
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, Netherlands
| | - Heshu Abdullah-Koolmees
- Division Laboratories, Pharmacy and Biomedical Genetics, Hospital Pharmacy, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Kathrin Blagec
- Centre for Medical Statistics, Informatics and Intelligent Systems, Institute of Artificial Intelligence, Medical University of Vienna, Vienna, Austria
| | - Tanja Blagus
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Stefan Böhringer
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, Netherlands; Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - Anne Cambon-Thomsen
- CNRS, Centre for Epidemiology and Research in Population health (CERPOP), Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Ka-Chun Cheung
- Medicines Information Centre, Royal Dutch Pharmacists Association (KNMP), The Hague, Netherlands
| | - Vera Hm Deneer
- Division Laboratories, Pharmacy and Biomedical Genetics, Hospital Pharmacy, University Medical Centre Utrecht, Utrecht, Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Netherlands
| | - Mathilde Dupui
- Service de pharmacologie médicale et clinique, CEIP-addictovigilance de Toulouse, faculté de médecine, CHU, Toulouse, France
| | | | - Siv Jonsson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Candace Joefield-Roka
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Katja S Just
- Institute of Clinical Pharmacology, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Lidija Konta
- Bio.logis Digital Health, Frankfurt am Main, Germany
| | - Rudolf Koopmann
- Bio.logis Digital Health, Frankfurt am Main, Germany; Diagnosticum Centre for Humangenetics, Frankfurt am Main, Germany
| | - Marjolein Kriek
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Saarbrücken, Germany
| | - Christina Mitropoulou
- The Golden Helix Foundation, London, UK; Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, Abu Dhabi, United Arab Emirates
| | | | - Victoria Rollinson
- Department of Pharmacology and Therapeutics, Wolfson Centre for Personalised Medicine, The University of Liverpool, Liverpool, UK
| | - Rossana Roncato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Matthias Samwald
- Centre for Medical Statistics, Informatics and Intelligent Systems, Institute of Artificial Intelligence, Medical University of Vienna, Vienna, Austria
| | - Elke Schaeffeler
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; iFIT Cluster of Excellence (EXC2180)-Image Guided and Functionally Instructed Tumour Therapies, University of Tuebingen, Tuebingen, Germany
| | - Maria Skokou
- University of Patras School of Health Sciences, Department of Pharmacy, Division of Pharmacology and Biosciences, Laboratory of Pharmacogenomics and Individualised Therapy, Patras, Greece
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; iFIT Cluster of Excellence (EXC2180)-Image Guided and Functionally Instructed Tumour Therapies, University of Tuebingen, Tuebingen, Germany; Department of Clinical Pharmacology, University of Tuebingen, Tuebingen, Germany; Department of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
| | - Daniela Steinberger
- Bio.logis Digital Health, Frankfurt am Main, Germany; Diagnosticum Centre for Humangenetics, Frankfurt am Main, Germany
| | - Julia C Stingl
- Institute of Clinical Pharmacology, University Hospital RWTH Aachen, Aachen, Germany
| | - Roman Tremmel
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Richard M Turner
- Department of Pharmacology and Therapeutics, Wolfson Centre for Personalised Medicine, The University of Liverpool, Liverpool, UK
| | - Mandy H van Rhenen
- Medicines Information Centre, Royal Dutch Pharmacists Association (KNMP), The Hague, Netherlands
| | - Cristina L Dávila Fajardo
- Clinical Pharmacy Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria Granada, Granada, Spain
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - George P Patrinos
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, Abu Dhabi, United Arab Emirates; Zayed Centre for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, Abu Dhabi, United Arab Emirates; University of Patras School of Health Sciences, Department of Pharmacy, Division of Pharmacology and Biosciences, Laboratory of Pharmacogenomics and Individualised Therapy, Patras, Greece; Erasmus University Medical Centre, Faculty of Medicine and Health Sciences, Department of Pathology-Clinical Bioinformatics Unit, Rotterdam, Netherlands
| | - Munir Pirmohamed
- Department of Pharmacology and Therapeutics, Wolfson Centre for Personalised Medicine, The University of Liverpool, Liverpool, UK
| | - Gere Sunder-Plassmann
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, Netherlands.
| |
Collapse
|
28
|
Keogh M, Fragala MS, Peter AP, Lorenz RA, Goldberg SE, Shaman JA. Early Insights From a Pharmacogenomic-Enriched Comprehensive Medication Management Program Implementation in an Adult Employee Population. J Occup Environ Med 2022; 64:e818-e822. [PMID: 36155954 PMCID: PMC9722373 DOI: 10.1097/jom.0000000000002705] [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: 02/04/2023]
Abstract
OBJECTIVES The aims of the study are to assess adoption of a pharmacogenomic-enriched comprehensive medication management program in a self-insured employer setting and to better understand medication risks that affect employees. METHODS Employees were identified to be at high risk of medication mismanagement and were subsequently provided with a program and process to improve their health. DNA testing, a clinical decision support system, and pharmacists were used to identify medication safety and effectiveness issues and to recommend appropriate changes. RESULTS A total of 10.6% of the invited employees enrolled in the program. Actionable recommendations were suggested by pharmacists for 85.8% of employees who completed the program, averaging 5.2 recommendations per person. CONCLUSIONS Implementation of a PGx + CMM program in a self-insured employer setting is feasible, detects risks in prescription regimens, and offers opportunities to improve medication management and reduce the burden of healthcare expenses.
Collapse
|
29
|
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
|