51
|
Scott SA, Scott ER, Seki Y, Chen AJ, Wallsten R, Owusu Obeng A, Botton MR, Cody N, Shi H, Zhao G, Brake P, Nicoletti P, Yang Y, Delio M, Shi L, Kornreich R, Schadt EE, Edelmann L. Development and Analytical Validation of a 29 Gene Clinical Pharmacogenetic Genotyping Panel: Multi-Ethnic Allele and Copy Number Variant Detection. Clin Transl Sci 2020; 14:204-213. [PMID: 32931151 PMCID: PMC7877843 DOI: 10.1111/cts.12844] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
To develop a novel pharmacogenetic genotyping panel, a multidisciplinary team evaluated available evidence and selected 29 genes implicated in interindividual drug response variability, including 130 sequence variants and additional copy number variants (CNVs). Of the 29 genes, 11 had guidelines published by the Clinical Pharmacogenetics Implementation Consortium. Targeted genotyping and CNV interrogation were accomplished by multiplex single‐base extension using the MassARRAY platform (Agena Biosciences) and multiplex ligation‐dependent probe amplification (MRC Holland), respectively. Analytical validation of the panel was accomplished by a strategic combination of > 500 independent tests performed on 170 unique reference material DNA samples, which included sequence variant and CNV accuracy, reproducibility, and specimen (blood, saliva, and buccal swab) controls. Among the accuracy controls were 32 samples from the 1000 Genomes Project that were selected based on their enrichment of sequence variants included in the pharmacogenetic panel (VarCover.org). Coupled with publicly available samples from the Genetic Testing Reference Materials Coordination Program (GeT‐RM), accuracy validation material was available for the majority (77%) of interrogated sequence variants (100% with average allele frequencies > 0.1%), as well as additional structural alleles with unique copy number signatures (e.g., CYP2D6*5, *13, *36, *68; CYP2B6*29; and CYP2C19*36). Accuracy and reproducibility for both genotyping and copy number were > 99.9%, indicating that the optimized panel platforms were precise and robust. Importantly, multi‐ethnic allele frequencies of the interrogated variants indicate that the vast majority of the general population carries at least one of these clinically relevant pharmacogenetic variants, supporting the implementation of this panel for pharmacogenetic research and/or clinical implementation programs.
Collapse
Affiliation(s)
- Stuart A Scott
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erick R Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | - Aniwaa Owusu Obeng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mariana R Botton
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Neal Cody
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | - Paola Nicoletti
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yao Yang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Lisong Shi
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth Kornreich
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Eric E Schadt
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lisa Edelmann
- Sema4, Stamford, Connecticut, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
52
|
Hoffman JM, Flynn AJ, Juskewitch JE, Freimuth RR. Biomedical Data Science and Informatics Challenges to Implementing Pharmacogenomics with Electronic Health Records. Annu Rev Biomed Data Sci 2020. [DOI: 10.1146/annurev-biodatasci-020320-093614] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pharmacogenomic information must be incorporated into electronic health records (EHRs) with clinical decision support in order to fully realize its potential to improve drug therapy. Supported by various clinical knowledge resources, pharmacogenomic workflows have been implemented in several healthcare systems. Little standardization exists across these efforts, however, which limits scalability both within and across clinical sites. Limitations in information standards, knowledge management, and the capabilities of modern EHRs remain challenges for the widespread use of pharmacogenomics in the clinic, but ongoing efforts are addressing these challenges. Although much work remains to use pharmacogenomic information more effectively within clinical systems, the experiences of pioneering sites and lessons learned from those programs may be instructive for other clinical areas beyond genomics. We present a vision of what can be achieved as informatics and data science converge to enable further adoption of pharmacogenomics in the clinic.
Collapse
Affiliation(s)
- James M. Hoffman
- Department of Pharmaceutical Sciences and the Office of Quality and Patient Care, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Allen J. Flynn
- Department of Learning Health Sciences, Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Justin E. Juskewitch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Robert R. Freimuth
- Division of Digital Health Sciences, Department of Health Sciences Research, Center for Individualized Medicine, and Information and Knowledge Management, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
53
|
Marrero RJ, Cicali EJ, Arwood MJ, Eddy E, DeRemer D, Ramnaraign BH, Daily KC, Jones D, Cook KJ, Cavallari LH, Wiisanen Weitzel K, Langaee T, Newsom KJ, Starostik P, Clare-Salzer MJ, Johnson JA, George TJ, Cooper-DeHoff RM. How to Transition from Single-Gene Pharmacogenetic Testing to Preemptive Panel-Based Testing: A Tutorial. Clin Pharmacol Ther 2020; 108:557-565. [PMID: 32460360 DOI: 10.1002/cpt.1912] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022]
Abstract
There have been significant advancements in precision medicine and approaches to medication selection based on pharmacogenetic results. With the availability of direct-to-consumer genetic testing and growing awareness of genetic interindividual variability, patient demand for more precise, individually tailored drug regimens is increasing. The University of Florida (UF) Health Precision Medicine Program (PMP) was established in 2011 to improve integration of genomic data into clinical practice. In the ensuing years, the UF Health PMP has successfully implemented several single-gene tests to optimize the precision of medication prescribing across a variety of clinical settings. Most recently, the UF Health PMP launched a custom-designed pharmacogenetic panel, including pharmacogenes relevant to supportive care medications commonly prescribed to patients undergoing chemotherapy treatment, referred to as "GatorPGx." This tutorial provides guidance and information to institutions on how to transition from the implementation of single-gene pharmacogenetic testing to a preemptive panel-based testing approach. Here, we demonstrate application of the preemptive panel in the setting of an adult solid tumor oncology clinic. Importantly, the information included herein can be applied to other clinical practice settings.
Collapse
Affiliation(s)
- Richard J Marrero
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Emily J Cicali
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Meghan J Arwood
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Elizabeth Eddy
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - David DeRemer
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | | | - Karen C Daily
- University of Florida Health Cancer Center, Gainesville, Florida, USA
| | - Dennie Jones
- University of Florida Health Cancer Center, Gainesville, Florida, USA
| | - Kelsey J Cook
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Larisa H Cavallari
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Kristin Wiisanen Weitzel
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Taimour Langaee
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Kimberly J Newsom
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Petr Starostik
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Michael J Clare-Salzer
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Thomas J George
- University of Florida Health Cancer Center, Gainesville, Florida, USA
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, University of Florida College of Pharmacy, Gainesville, Florida, USA
| |
Collapse
|
54
|
L Rogers S, Keeling NJ, Giri J, Gonzaludo N, Jones JS, Glogowski E, Formea CM. PARC report: a health-systems focus on reimbursement and patient access to pharmacogenomics testing. Pharmacogenomics 2020; 21:785-796. [DOI: 10.2217/pgs-2019-0192] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pharmacogenomics test coverage and reimbursement are major obstacles to clinical uptake. Several early adopter programs have been successfully initiated through dedicated investments by federal and institutional research funding. As a result of research endeavors, evidence has grown sufficiently to support development of pharmacogenomics guidelines. However, clinical uptake is still limited. Third-party payer support plays an important role in increasing adoption, which to date has been limited to reactive single-gene testing. Access to and interest in direct-to-consumer genetic testing are driving demand for increasing healthcare providers and third-party awareness of this burgeoning field. Pharmacogenomics implementation models developed by early adopters promise to expand patient access and options, as testing continues to increase due to growing consumer interest and falling test prices.
Collapse
Affiliation(s)
- Sara L Rogers
- American Society of Pharmacovigilance, PO Box 20433, Houston, TX 77225, USA
| | - Nicholas J Keeling
- Department of Pharmacy Administration, The University of Mississippi School of Pharmacy, 223 Faser Hall, MS 38677, USA
| | - Jyothsna Giri
- Center for Individualized Medicine, Mayo Clinic, 200 First Street SW, MN 55905, USA
| | - Nina Gonzaludo
- Illumina, Inc., 200 Lincoln Centre Drive, Foster City, CA 94404, USA
| | - J Shawn Jones
- Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, 5920 Forest Park Rd, Suite 500, Dallas, TX 75235, USA
| | | | - Christine M Formea
- Center for Individualized Medicine, Mayo Clinic, 200 First Street SW, MN 55905, USA
- Department of Pharmacy Services & Intermountain Precision Genomics, Intermountain Healthcare Pharmacy Services, 4393 S. Riverboat Road, Taylorsville, UT 84123, USA
| |
Collapse
|
55
|
Mizuno T, Dong M, Taylor ZL, Ramsey LB, Vinks AA. Clinical implementation of pharmacogenetics and model-informed precision dosing to improve patient care. Br J Clin Pharmacol 2020; 88:1418-1426. [PMID: 32529759 DOI: 10.1111/bcp.14426] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Providing maximal therapeutic efficacy without toxicity is a universal goal of rational drug therapy. However, substantial between-patient variability in drug response often impedes such successful treatments and brings the necessity of tailoring drug dose to individual needs for more precise therapy. In many cases plenty of patient characteristics, such as body size, genetic makeup and environmental factors, need to be taken into consideration to find the optimal dose in clinical practice. A pharmacokinetics and pharmacodynamics (PK/PD) model-informed approach offers integration of various patient information to provide an expectation of drug response and derive practical dose estimates to support clinicians' dosing decisions. Such an approach was pioneered in the late 1970s, but its broad clinical acceptance and implementation have been hampered by the lack of widespread computer technology, including user-friendly software tools. This has significantly changed in recent years. With the advent of electronic health records (EHRs) and the ubiquity of user-friendly software tools, we now experience a convergence of clinical information, pharmacogenetics, systems pharmacology and pharmacometrics, and technology. Advanced pharmacometrics research is now more appliable and implementable to improve health care. This article presents examples of successful development and implementation of pharmacogenetics-guided and PK/PD model-informed decision support to facilitate precision dosing, including the development of an EHR-embedded decision support tool. Through the integration of clinical decision support tools in EHRs, clinical pharmacometrics support can be brought directly to the clinical team and the bedside.
Collapse
Affiliation(s)
- Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zachary L Taylor
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Molecular, Cellular, and Biochemical Pharmacology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Laura B Ramsey
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
56
|
Determinants of stakeholders' intention to adopt pharmacogenomic. THE PHARMACOGENOMICS JOURNAL 2020; 20:801-812. [PMID: 32457399 DOI: 10.1038/s41397-020-0167-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/08/2020] [Accepted: 05/14/2020] [Indexed: 12/28/2022]
Abstract
Pharmacogenomics (PGx) testing, which aims to identify the genes that affect our responses to drugs, has been favoured by healthcare professionals as a means of maximising drug efficacy and improving the safety and cost-effectiveness of healthcare. Support from the public is needed to determine the successful development of this technology and its implementation in society. Therefore, the objective of this paper was to analyse factors that influence stakeholders' intentions to adopt pharmacogenomic testing in Malaysia. A validated instrument was administered through face-to-face interviews with a total of 421 adult respondents who were stratified according to 2 stakeholder groups: healthcare providers (n = 221) and patients/family members (n = 200). The data were then analysed using SPSS® version 24 software and the advanced multivariate statistical approach of Partial Least Square (PLS) path modelling in order to analyse the complex relationships among variables. Results of the studies indicated that the Malaysian stakeholders had a high amount of trust in the key players (mean score of 5.31), perceived high benefits (mean score of 5.53) and claimed to have high intentions of adopting PGx (mean score of 5.39). The majority of the predictors have significant direct relationships with the intention to adopt PGx, with the exception of moral concerns. Perceived benefits appeared to be the most important direct predictor of the intention to adopt PGx testing (ß = 0.371, P < 0.001) followed by trust in the key players (ß = 0.312, P < 0.001), engagement (ß = 0.272, P < 0.001) and religiosity (ß = 0.133, P < 0.01). In addition, perceived risks also had a direct negative association with the intention to adopt PGx (ß = -0.096, P < 0.05). At the same time, the perceived benefits also served as a mediator for all the other factors except risk. The results provide insights into the multidimensional nature of the determinants of the intention to adopt PGx testing in Malaysia. Although the results showed that the stakeholders in Malaysia were very positive towards PGx testing, they were also cautious about it. The predictors identified in this study can serve as indicators for social acceptance of PGx testing to facilitate the clinical research and implementation of PGx.
Collapse
|
57
|
Karas Kuželički N, Prodan Žitnik I, Gurwitz D, Llerena A, Cascorbi I, Siest S, Simmaco M, Ansari M, Pazzagli M, Di Resta C, Brandslund I, Schwab M, Vermeersch P, Lunshof JE, Dedoussis G, Flordellis CS, Fuhr U, Stingl JC, van Schaik RH, Manolopoulos VG, Marc J. Pharmacogenomics education in medical and pharmacy schools: conclusions of a global survey. Pharmacogenomics 2020; 20:643-657. [PMID: 31250730 DOI: 10.2217/pgs-2019-0009] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aim: The need for pharmacogenomic education is becoming more and more urgent. Our aim was to evaluate the progress in pharmacogenomics education since then, and to put forward further recommendations. Methods: A survey was sent to 248 schools of medicine, pharmacy, nursing and health professions around the world. Results: The majority of the study programs (87%) include pharmacogenomics education, which is generally taught as part of the pharmacology curriculum. On average, educators and teachers have selected appropriate and highly relevant pharmacogenomics biomarkers to include in their teaching programs. Conclusions: Based on the results, we can conclude that the state of pharmacogenomics education at the surveyed universities has improved substantially since 2005.
Collapse
Affiliation(s)
| | | | - David Gurwitz
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | - Sofia Siest
- INSERM UMR U1122, University of Loraine, Nancy, France
| | | | - Marc Ansari
- Onco-Hematology Unit, University Hospital of Geneva, Geneva, Switzerland.,Cansearch Research Laboratory, Geneva Medical School, Geneva, Switzerland
| | | | - Chiara Di Resta
- Vita-Salute San Raffaele University, Milan, Italy.,Genomics Unit for Diagnosis of Human Genetics, Division of Genetics & Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ivan Brandslund
- University of Southern Denmark, Vejle Hospital, Vejle, Denmark
| | - Matthias Schwab
- Department of Clinical Pharmacology, University Hospital Tuebingen, Tuebingen, Germany.,Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Pharmacy & Biochemistry, University of Tuebingen, Tuebingen, Germany
| | | | - Jeantine E Lunshof
- University of Gröningen, University Medical Center Gröningen, Gröningen, The Netherlands.,Harvard Medical School, Boston, MA, USA.,Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | - Uwe Fuhr
- Centre of Pharmacology, University of Cologne, Cologne, Germany
| | | | - Ron Hn van Schaik
- Deptartment of Clinical Chemistry, Erasmus MC Rotterdam, Rotterdam, The Netherlands.,European Society of Pharmacogenomics & Personalised Therapy, Via Carlo Farini 81, Milan, Italy
| | - Vangelis G Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Janja Marc
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | | |
Collapse
|
58
|
Smith DM, Peshkin BN, Springfield TB, Brown RP, Hwang E, Kmiecik S, Shapiro R, Eldadah Z, Lundergan C, McAlduff J, Levin B, Swain SM. Pharmacogenetics in Practice: Estimating the Clinical Actionability of Pharmacogenetic Testing in Perioperative and Ambulatory Settings. Clin Transl Sci 2020; 13:618-627. [PMID: 31961467 PMCID: PMC7214646 DOI: 10.1111/cts.12748] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/17/2019] [Indexed: 01/04/2023] Open
Abstract
Most literature describing pharmacogenetic implementations are within academic medical centers and use single-gene tests. Our objective was to describe the results and lessons learned from a multisite pharmacogenetic pilot that utilized panel-based testing in academic and nonacademic settings. This was a retrospective analysis of 667 patients from a pilot in 4 perioperative and 5 outpatient cardiology clinics. Recommendations related to 12 genes and 65 drugs were classified as actionable or not actionable. They were ascertained from Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines and US Food and Drug Administration (FDA) labeling. Patients displayed a high prevalence of actionable results (88%, 99%) and use of medications (28%, 46%) with FDA or CPIC recommendations, respectively. Sixteen percent of patients had an actionable result for a current medication per CPIC compared with 5% per FDA labeling. A systematic approach by a health system may be beneficial given the quantity and diversity of patients affected.
Collapse
Affiliation(s)
- D. Max Smith
- MedStar HealthColumbiaMarylandUSA
- Georgetown University Medical CenterWashingtonDCUSA
| | | | | | | | | | | | | | - Zayd Eldadah
- MedStar Washington Hospital CenterWashingtonDCUSA
| | | | | | | | - Sandra M. Swain
- MedStar HealthColumbiaMarylandUSA
- Georgetown University Medical CenterWashingtonDCUSA
| |
Collapse
|
59
|
Chan CWH, Law BMH, So WKW, Chow KM, Waye MMY. Pharmacogenomics of breast cancer: highlighting CYP2D6 and tamoxifen. J Cancer Res Clin Oncol 2020; 146:1395-1404. [PMID: 32270286 DOI: 10.1007/s00432-020-03206-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To review recent pharmacogenomics studies on breast cancer patients undergoing tamoxifen therapy, highlighting how our knowledge on cytochrome P450 2D6 (CYP2D6) can help to guide the development of adjuvant therapies for these patients. METHODS A comprehensive literature search was conducted. Articles reporting findings pertaining to the effect of CYP2D6 on the therapeutic efficacy of tamoxifen, those reporting how targeting CYP2D6 could inform tamoxifen-based therapy development, and those on the tamoxifen effects on cell lines and animal models were included in the review. RESULTS With CYP2D6 being the primary enzyme for tamoxifen metabolism, single-nucleotide polymorphisms (SNPs) in this gene were one of the determinants in the rate of tamoxifen metabolism, thereby potentially having an effect on the efficacy of tamoxifen-based therapies. Our review indicates the potential effectiveness of targeting these SNPs, including those for the CYP2D6*10 allele (c. 100C > T), in modifying the level of tamoxifen metabolism. These findings suggest the importance of pharmacogenomics research in our understanding of the efficacy of adjuvant therapies. However, the involvement of multiple enzymes in tamoxifen metabolism, dietary factors, ethnic differences in gene frequencies, and patients' compliance to tamoxifen therapies in studies do present challenges in pharmacogenomics research. CONCLUSIONS Pharmacogenomics could play important roles in mediating the advancement of tamoxifen-based adjuvant therapies. Research efforts should be directed towards the exploration of further SNPs of CYP2D6 that affect tamoxifen metabolism, as well as epigenetic changes in CYP2D6, enabling the design of precision medicine and confirming clinical validity in the use of pharmacogenomics for tamoxifen.
Collapse
Affiliation(s)
- Carmen W H Chan
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, 6/F Esther Lee Building, Hong Kong, China
| | - Bernard M H Law
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, 6/F Esther Lee Building, Hong Kong, China
| | - Winnie K W So
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, 6/F Esther Lee Building, Hong Kong, China
| | - Ka Ming Chow
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, 6/F Esther Lee Building, Hong Kong, China
| | - Mary M Y Waye
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, 6/F Esther Lee Building, Hong Kong, China. .,The Croucher Laboratory for Human Genomics, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
60
|
Kostyuk GP, Zakharova NV, Reznik AM, Surkova EI, Ilinsky VV. [Perspectives of the use of pharmacogenetic tests in neurology and psychiatry]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 119:131-135. [PMID: 31626230 DOI: 10.17116/jnevro2019119091131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The review is devoted to the analysis of the current state of pharmacogenetic research and their use in psychiatric practice. The main genes responsible for the pharmacodynamics and pharmacokinetics of drugs used in psychiatry are listed. Foreign pharmacogenetic clinical recommendations and progress on their implementation in medical practice in various countries of Europe and the USA are analyzed. The need to create Russian clinical guidelines on pharmacogenomics to improve the effectiveness of patient care and to implement a personalized approach to therapy is discussed.
Collapse
Affiliation(s)
- G P Kostyuk
- Alekseev Psychiatric Clinical Hospital #1, Moscow, Russia
| | - N V Zakharova
- Alekseev Psychiatric Clinical Hospital #1, Moscow, Russia
| | - A M Reznik
- Medical Institute of Ongoing Education of 'Moscow National University of Food Production', Moscow, Russia
| | | | | |
Collapse
|
61
|
Hernandez W, Danahey K, Pei X, Yeo KTJ, Leung E, Volchenboum SL, Ratain MJ, Meltzer DO, Stranger BE, Perera MA, O'Donnell PH. Pharmacogenomic genotypes define genetic ancestry in patients and enable population-specific genomic implementation. THE PHARMACOGENOMICS JOURNAL 2020; 20:126-135. [PMID: 31506565 PMCID: PMC7184888 DOI: 10.1038/s41397-019-0095-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/02/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022]
Abstract
The importance of genetic ancestry characterization is increasing in genomic implementation efforts, and clinical pharmacogenomic guidelines are being published that include population-specific recommendations. Our aim was to test the ability of focused clinical pharmacogenomic SNP panels to estimate individual genetic ancestry (IGA) and implement population-specific pharmacogenomic clinical decision-support (CDS) tools. Principle components and STRUCTURE were utilized to assess differences in genetic composition and estimate IGA among 1572 individuals from 1000 Genomes, two independent cohorts of Caucasians and African Americans (AAs), plus a real-world validation population of patients undergoing pharmacogenomic genotyping. We found that clinical pharmacogenomic SNP panels accurately estimate IGA compared to genome-wide genotyping and identify AAs with ≥70 African ancestry (sensitivity >82%, specificity >80%, PPV >95%, NPV >47%). We also validated a new AA-specific warfarin dosing algorithm for patients with ≥70% African ancestry and implemented it at our institution as a novel CDS tool. Consideration of IGA to develop an institutional CDS tool was accomplished to enable population-specific pharmacogenomic guidance at the point-of-care. These capabilities were immediately applied for guidance of warfarin dosing in AAs versus Caucasians, but also provide a real-world model that can be extended to other populations and drugs as actionable genomic evidence accumulates.
Collapse
Affiliation(s)
- Wenndy Hernandez
- University of Chicago, Department of Medicine, Section of Genetic Medicine, Section of Cardiology, Chicago, IL, USA
| | - Keith Danahey
- University of Chicago, Center for Personalized Therapeutics, Chicago, IL, USA
- University of Chicago, Center for Research Informatics, Chicago, IL, USA
| | - Xun Pei
- University of Chicago, Center for Personalized Therapeutics, Chicago, IL, USA
- University of Chicago, Department of Pathology, UChicago Advanced Technology Clinical Pharmacogenomics Laboratory, Chicago, IL, USA
| | - Kiang-Teck J Yeo
- University of Chicago, Department of Pathology, UChicago Advanced Technology Clinical Pharmacogenomics Laboratory, Chicago, IL, USA
| | - Edward Leung
- University of Chicago, Department of Pathology, UChicago Advanced Technology Clinical Pharmacogenomics Laboratory, Chicago, IL, USA
- University of Southern California, Keck School of Medicine, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
| | | | - Mark J Ratain
- University of Chicago, Center for Personalized Therapeutics, Chicago, IL, USA
- University of Chicago, Department of Medicine, Chicago, IL, USA
- University of Chicago, Committee on Clinical Pharmacology and Pharmacogenomics, Chicago, IL, USA
| | - David O Meltzer
- University of Chicago, Department of Medicine, Chicago, IL, USA
| | - Barbara E Stranger
- University of Chicago, Department of Medicine, Section of Genetic Medicine, Section of Cardiology, Chicago, IL, USA
- University of Chicago, Institute of Genomics and Systems Biology, and Center for Data Intensive Science, Chicago, IL, USA
| | - Minoli A Perera
- Northwestern University, Department of Pharmacology, Chicago, IL, USA
| | - Peter H O'Donnell
- University of Chicago, Center for Personalized Therapeutics, Chicago, IL, USA.
- University of Chicago, Department of Medicine, Chicago, IL, USA.
- University of Chicago, Committee on Clinical Pharmacology and Pharmacogenomics, Chicago, IL, USA.
| |
Collapse
|
62
|
Rahawi S, Naik H, Blake KV, Owusu Obeng A, Wasserman RM, Seki Y, Funanage VL, Oishi K, Scott SA. Knowledge and attitudes on pharmacogenetics among pediatricians. J Hum Genet 2020; 65:437-444. [PMID: 31983733 DOI: 10.1038/s10038-020-0723-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/06/2020] [Accepted: 01/06/2020] [Indexed: 11/09/2022]
Abstract
Increasing enthusiasm for clinical pharmacogenetic testing and the availability of pharmacogenetic-based guidelines indicate that pediatricians will increasingly be expected to interpret and apply pharmacogenetic test results into medical care. Previous studies have identified a lack of knowledge on pharmacogenetics across many physician specialties; however, this has not been systematically assessed among pediatricians. To evaluate pediatrician knowledge, attitude, and educational interest in pharmacogenetics, we surveyed physician cohorts from both the United States (U.S.) and Japan. A total of 282 pediatricians (210 from the U.S. and 72 from Japan) participated in an anonymous survey (online or hardcopy) on pharmacogenetics knowledge, perception, and education. Over 50% of all respondents had >10 years of clinical experience and >75% had some prior education in genetics. However, <10% felt they were familiar with pharmacogenetics, which was very consistent with <20% of the U.S. pediatricians correctly responding to a codeine/CYP2D6 pharmacogenetics knowledge question and <10% of U.S. pediatricians being aware of the Clinical Pharmacogenetics Implementation Consortium (CPIC). Despite being generally unfamiliar with pharmacogenetics, >80% of all respondents indicated that implementation of clinical pharmacogenetic testing will improve efficacy and safety, and that pediatricians should be capable of applying this testing to their practice. Moreover, the majority (83.1%) were interested in educational opportunities on pharmacogenetics, particularly on result interpretation and therapeutic recommendations. Taken together, these data indicate that although practical knowledge of pharmacogenetics among pediatricians in the U.S. and Japan is currently very low, their interest in clinical pharmacogenetics and related education is high, which will likely facilitate future implementation.
Collapse
Affiliation(s)
- Shahad Rahawi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sema4, Stamford, CT, 06902, USA
| | - Hetanshi Naik
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kathryn V Blake
- Department of Biomedical Research, Nemours Children's Health System, Wilmington, DE, USA.,Department of Biomedical Research, Nemours Children's Health System, Jacksonville, Orlando, FL, USA
| | - Aniwaa Owusu Obeng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Pharmacy, The Mount Sinai Hospital, New York, NY, 10029, USA
| | - Rachel M Wasserman
- Department of Biomedical Research, Nemours Children's Health System, Wilmington, DE, USA.,Department of Biomedical Research, Nemours Children's Health System, Jacksonville, Orlando, FL, USA
| | - Yoshinori Seki
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sema4, Stamford, CT, 06902, USA
| | - Vicky L Funanage
- Department of Biomedical Research, Nemours Children's Health System, Wilmington, DE, USA.,Department of Biomedical Research, Nemours Children's Health System, Jacksonville, Orlando, FL, USA
| | - Kimihiko Oishi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stuart A Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Sema4, Stamford, CT, 06902, USA.
| |
Collapse
|
63
|
Applying Pharmacogenomics to Antifungal Selection and Dosing: Are We There Yet? CURRENT FUNGAL INFECTION REPORTS 2020; 14:63-75. [PMID: 32256938 DOI: 10.1007/s12281-020-00371-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Purpose of Review This review summarizes recent literature for applying pharmacogenomics to antifungal selection and dosing, providing an approach to implementing antifungal pharmacogenomics in clinical practice. Recent Findings The Clinical Pharmacogenetics Implementation Consortium published guidelines on CYP2C19 and voriconazole, with recommendations to use alternative antifungals or adjust voriconazole dose with close therapeutic drug monitoring (TDM). Recent studies demonstrate an association between CYP2C19 phenotype and voriconazole levels, clinical outcomes, and adverse events. Additionally, CYP2C19-guided preemptive dose adjustment demonstrated benefit in two prospective studies for prophylaxis. Pharmacokinetic-pharmacodynamic modeling studies have generated proposed voriconazole treatment doses based on CYP2C19 phenotypes, with further validation studies needed. Summary Sufficient evidence is available for implementing CYP2C19-guided voriconazole selection and dosing among select patients at risk for invasive fungal infections. The institution needs appropriate infrastructure for pharmacogenomic testing, integration of results in the clinical decision process, with TDM confirmation of goal trough achievement, to integrate antifungal pharmacogenomics into routine clinical care.
Collapse
|
64
|
El Rouby N, Alrwisan A, Langaee T, Lipori G, Angiolillo DJ, Franchi F, Riva A, Elsey A, Johnson JA, Cavallari LH, Winterstein AG. Clinical Utility of Pharmacogene Panel-Based Testing in Patients Undergoing Percutaneous Coronary Intervention. Clin Transl Sci 2020; 13:473-481. [PMID: 31758664 PMCID: PMC7214641 DOI: 10.1111/cts.12729] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/02/2019] [Indexed: 12/19/2022] Open
Abstract
We aimed to estimate the utility of panel‐based pharmacogenetic testing of patients undergoing percutaneous coronary intervention (PCI). Utilization of Clinical Pharmacogenetic Implementation Consortium (CPIC) level A/B drugs after PCI was estimated in a national sample of IBM MarketScan beneficiaries. Genotype data from University of Florida (UF) patients (n = 211) who underwent PCI were used to project genotype‐guided opportunities among MarketScan beneficiaries with at least one (N = 105,547) and five (N = 12,462) years of follow‐up data. The actual incidence of genotype‐guided prescribing opportunities was determined among UF patients. In MarketScan, 50.0% (52,799/105,547) over 1 year and 68.0% (8,473/12,462) over 5 years had ≥ 1 CPIC A/B drug besides antiplatelet therapy prescribed, with a projected incidence of genotype‐guided prescribing opportunities of 39% at 1 year and 52% at 5 years. Genotype‐guided prescribing opportunities occurred in 32% of UF patients. Projected and actual incidence of genotype‐guided opportunities among two cohorts supports the utility of panel‐based testing among patients who underwent PCI.
Collapse
Affiliation(s)
- Nihal El Rouby
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Adel Alrwisan
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Taimour Langaee
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Gloria Lipori
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,UF Health & UF Health Sciences Center, Gainesville, Florida, USA
| | - Dominick J Angiolillo
- Division of Cardiology, Department of Medicine, University of Florida, Jacksonville, Florida, USA
| | - Francesco Franchi
- Division of Cardiology, Department of Medicine, University of Florida, Jacksonville, Florida, USA
| | - Alberto Riva
- Bioinformatics Core, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, USA
| | - Amanda Elsey
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Larisa H Cavallari
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Almut G Winterstein
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Epidemiology, University of Florida, Gainesville, Florida, USA.,Center for Drug Evaluation and Safety, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
65
|
Herr TM, Peterson JF, Rasmussen LV, Caraballo PJ, Peissig PL, Starren JB. Pharmacogenomic clinical decision support design and multi-site process outcomes analysis in the eMERGE Network. J Am Med Inform Assoc 2020; 26:143-148. [PMID: 30590574 DOI: 10.1093/jamia/ocy156] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/05/2018] [Indexed: 11/12/2022] Open
Abstract
To better understand the real-world effects of pharmacogenomic (PGx) alerts, this study aimed to characterize alert design within the eMERGE Network, and to establish a method for sharing PGx alert response data for aggregate analysis. Seven eMERGE sites submitted design details and established an alert logging data dictionary. Six sites participated in a pilot study, sharing alert response data from their electronic health record systems. PGx alert design varied, with some consensus around the use of active, post-test alerts to convey Clinical Pharmacogenetics Implementation Consortium recommendations. Sites successfully shared response data, with wide variation in acceptance and follow rates. Results reflect the lack of standardization in PGx alert design. Standards and/or larger studies will be necessary to fully understand PGx impact. This study demonstrated a method for sharing PGx alert response data and established that variation in system design is a significant barrier for multi-site analyses.
Collapse
Affiliation(s)
- Timothy M Herr
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Josh F Peterson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Luke V Rasmussen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Pedro J Caraballo
- Department of Medicine and Center for Translational Informatics and Knowledge Management, Mayo Clinic, Rochester, Minnesota, USA
| | - Peggy L Peissig
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Justin B Starren
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
66
|
Truong TM, Lipschultz E, Danahey K, Schierer E, Ratain MJ, O'Donnell PH. Assessment of Patient Knowledge and Perceptions of Pharmacogenomics Before and After Using a Mock Results Patient Web Portal. Clin Transl Sci 2020; 13:78-87. [PMID: 31490020 PMCID: PMC6951853 DOI: 10.1111/cts.12681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/03/2019] [Indexed: 01/28/2023] Open
Abstract
Our objective was to build a mock pharmacogenomic (PGx) patient portal and assess its ability to disseminate test results and information to patients. The YourPGx Portal delivered four sample PGx results (omeprazole, simvastatin, clopidogrel, and codeine). We hosted two study groups to assess patient knowledge and perceptions of PGx before and after accessing the portal. Ten PGx-tested and 10 traditional care participants were included (average 61 years, 60% women, 50% African American, and 55% had a bachelor's/advanced degree). Participants scored significantly higher on the post-test compared with the pre-test, with no significant differences between baseline scores or score change between the groups. Patient perceptions also improved after accessing the portal-more patients wanted their providers to have access to test results, and more patients would encourage family/friends to get PGx testing. Patients would share their test results with their healthcare providers, spouse/partner, and family; none would share results with their friends or social media. Almost all patients (95%) said the portal was easy to use and 65% said it was easy to understand. In this pilot study, patients' knowledge and perceptions of PGx improved after accessing the YourPGx Portal.
Collapse
Affiliation(s)
- Tien M. Truong
- Committee on Clinical Pharmacology and PharmacogenomicsThe University of ChicagoChicagoIllinoisUSA
- Center for Personalized TherapeuticsThe University of ChicagoChicagoIllinoisUSA
| | - Elizabeth Lipschultz
- Center for Personalized TherapeuticsThe University of ChicagoChicagoIllinoisUSA
- Center for Research InformaticsThe University of ChicagoChicagoIllinoisUSA
| | - Keith Danahey
- Center for Personalized TherapeuticsThe University of ChicagoChicagoIllinoisUSA
- Center for Research InformaticsThe University of ChicagoChicagoIllinoisUSA
| | - Emily Schierer
- Center for Personalized TherapeuticsThe University of ChicagoChicagoIllinoisUSA
| | - Mark J. Ratain
- Committee on Clinical Pharmacology and PharmacogenomicsThe University of ChicagoChicagoIllinoisUSA
- Center for Personalized TherapeuticsThe University of ChicagoChicagoIllinoisUSA
- Department of MedicineThe University of ChicagoChicagoIllinoisUSA
| | - Peter H. O'Donnell
- Committee on Clinical Pharmacology and PharmacogenomicsThe University of ChicagoChicagoIllinoisUSA
- Center for Personalized TherapeuticsThe University of ChicagoChicagoIllinoisUSA
- Department of MedicineThe University of ChicagoChicagoIllinoisUSA
| |
Collapse
|
67
|
Fortinguerra S, Sorrenti V, Giusti P, Zusso M, Buriani A. Pharmacogenomic Characterization in Bipolar Spectrum Disorders. Pharmaceutics 2019; 12:E13. [PMID: 31877761 PMCID: PMC7022469 DOI: 10.3390/pharmaceutics12010013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/14/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022] Open
Abstract
The holistic approach of personalized medicine, merging clinical and molecular characteristics to tailor the diagnostic and therapeutic path to each individual, is steadily spreading in clinical practice. Psychiatric disorders represent one of the most difficult diagnostic challenges, given their frequent mixed nature and intrinsic variability, as in bipolar disorders and depression. Patients misdiagnosed as depressed are often initially prescribed serotonergic antidepressants, a treatment that can exacerbate a previously unrecognized bipolar condition. Thanks to the use of the patient's genomic profile, it is possible to recognize such risk and at the same time characterize specific genetic assets specifically associated with bipolar spectrum disorder, as well as with the individual response to the various therapeutic options. This provides the basis for molecular diagnosis and the definition of pharmacogenomic profiles, thus guiding therapeutic choices and allowing a safer and more effective use of psychotropic drugs. Here, we report the pharmacogenomics state of the art in bipolar disorders and suggest an algorithm for therapeutic regimen choice.
Collapse
Affiliation(s)
- Stefano Fortinguerra
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; (S.F.); (V.S.)
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| | - Vincenzo Sorrenti
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; (S.F.); (V.S.)
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
- Bendessere™ Study Center, Solgar Italia Multinutrient S.p.A., 35131 Padova, Italy
| | - Pietro Giusti
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| | - Morena Zusso
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| | - Alessandro Buriani
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; (S.F.); (V.S.)
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| |
Collapse
|
68
|
Truong TM, Apfelbaum J, Shahul S, Anitescu M, Danahey K, Knoebel RW, Liebovitz D, Karrison T, van Wijk XMR, Yeo KTJ, Meltzer D, Ratain MJ, O'Donnell PH. The ImPreSS Trial: Implementation of Point-of-Care Pharmacogenomic Decision Support in Perioperative Care. Clin Pharmacol Ther 2019; 106:1179-1183. [PMID: 31433489 PMCID: PMC7214100 DOI: 10.1002/cpt.1567] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/29/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Tien M Truong
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
| | - Jeffrey Apfelbaum
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, USA
| | - Sajid Shahul
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, USA
| | - Magdalena Anitescu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, USA
| | - Keith Danahey
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
- Center for Research Informatics, The University of Chicago, Chicago, Illinois, USA
| | - Randall W Knoebel
- Department of Pharmacy, The University of Chicago, Chicago, Illinois, USA
| | - David Liebovitz
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Theodore Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, Illinois, USA
| | - Xander M R van Wijk
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Kiang-Teck J Yeo
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - David Meltzer
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Mark J Ratain
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Peter H O'Donnell
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
69
|
Pharmacogenes (PGx-genes): Current understanding and future directions. Gene 2019; 718:144050. [DOI: 10.1016/j.gene.2019.144050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022]
|
70
|
Fuselli S. Beyond drugs: the evolution of genes involved in human response to medications. Proc Biol Sci 2019; 286:20191716. [PMID: 31640517 DOI: 10.1098/rspb.2019.1716] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The genetic variation of our species reflects human demographic history and adaptation to diverse local environments. Part of this genetic variation affects individual responses to exogenous substances, such as food, pollutants and drugs, and plays an important role in drug efficacy and safety. This review provides a synthesis of the evolution of loci implicated in human pharmacological response and metabolism, interpreted within the theoretical framework of population genetics and molecular evolution. In particular, I review and discuss key evolutionary aspects of different pharmacogenes in humans and other species, such as the relationship between the type of substrates and rate of evolution; the selective pressure exerted by landscape variables or dietary habits; expected and observed patterns of rare genetic variation. Finally, I discuss how this knowledge can be translated directly or after the implementation of specific studies, into practical guidelines.
Collapse
Affiliation(s)
- Silvia Fuselli
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
71
|
Roden DM, Van Driest SL, Wells QS, Mosley JD, Denny JC, Peterson JF. Opportunities and Challenges in Cardiovascular Pharmacogenomics: From Discovery to Implementation. Circ Res 2019; 122:1176-1190. [PMID: 29700066 DOI: 10.1161/circresaha.117.310965] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review will provide an overview of the principles of pharmacogenomics from basic discovery to implementation, encompassing application of tools of contemporary genome science to the field (including areas of apparent divergence from disease-based genomics), a summary of lessons learned from the extensively studied drugs clopidogrel and warfarin, the current status of implementing pharmacogenetic testing in practice, the role of genomics and related tools in the drug development process, and a summary of future opportunities and challenges.
Collapse
Affiliation(s)
- Dan M Roden
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.) .,Department of Pharmacology (D.M.R., Q.S.W.).,Department of Biomedical Informatics (D.M.R., J.C.D., J.F.P.)
| | - Sara L Van Driest
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Pediatrics (S.L.V.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Quinn S Wells
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Pharmacology (D.M.R., Q.S.W.)
| | - Jonathan D Mosley
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.)
| | - Joshua C Denny
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Biomedical Informatics (D.M.R., J.C.D., J.F.P.)
| | - Josh F Peterson
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Biomedical Informatics (D.M.R., J.C.D., J.F.P.)
| |
Collapse
|
72
|
Suzuki O, Dong OM, Howard RM, Wiltshire T. Characterizing the pharmacogenome using molecular inversion probes for targeted next-generation sequencing. Pharmacogenomics 2019; 20:1005-1020. [DOI: 10.2217/pgs-2019-0057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Aim: This study assesses the technical performance and cost of a targeted next-generation sequencing (NGS) multigene pharmacogenetic (PGx) test. Materials & methods: A genetic test was developed for 21 PGx genes using molecular inversion probes to generate library fragments for NGS. Performance of this test was assessed using 53 unique reference control cell lines from the Genetic Testing Reference Materials Coordination Program (GeT-RM). Results: 93.7% of variants were successfully called and the repeatability rate was 99.9%. Reference calls were available for 78.4% of diplotype calls resulting from PGx testing, and concordance for the test was 85.7%. Cost per sample was $32–$56. Conclusion: A targeted NGS assay using molecular inversion probe technology is able to characterize the pharmacogenome efficiently.
Collapse
Affiliation(s)
- Oscar Suzuki
- Division of Pharmacotherapy & Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Olivia M Dong
- Division of Pharmacotherapy & Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel M Howard
- Division of Pharmacotherapy & Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tim Wiltshire
- Division of Pharmacotherapy & Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
73
|
Krebs K, Milani L. Translating pharmacogenomics into clinical decisions: do not let the perfect be the enemy of the good. Hum Genomics 2019; 13:39. [PMID: 31455423 PMCID: PMC6712791 DOI: 10.1186/s40246-019-0229-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
The field of pharmacogenomics (PGx) is gradually shifting from the reactive testing of single genes toward the proactive testing of multiple genes to improve treatment outcomes, reduce adverse events, and decrease the burden of unnecessary costs for healthcare systems. Despite the progress in the field of pharmacogenomics, its implementation into routine care has been slow due to several barriers. However, in recent years, the number of studies on the implementation of PGx has increased, all providing a wealth of knowledge on different solutions for overcoming the obstacles that have been emphasized over the past years. This review focuses on some of the challenges faced by these initiatives, the solutions and different approaches for testing that they suggest, and the evidence that they provide regarding the benefits of preemptive PGx testing.
Collapse
Affiliation(s)
- Kristi Krebs
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
74
|
Hippman C, Nislow C. Pharmacogenomic Testing: Clinical Evidence and Implementation Challenges. J Pers Med 2019; 9:jpm9030040. [PMID: 31394823 PMCID: PMC6789586 DOI: 10.3390/jpm9030040] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/23/2019] [Accepted: 08/02/2019] [Indexed: 12/25/2022] Open
Abstract
Pharmacogenomics can enhance patient care by enabling treatments tailored to genetic make-up and lowering risk of serious adverse events. As of June 2019, there are 132 pharmacogenomic dosing guidelines for 99 drugs and pharmacogenomic information is included in 309 medication labels. Recently, the technology for identifying individual-specific genetic variants (genotyping) has become more accessible. Next generation sequencing (NGS) is a cost-effective option for genotyping patients at many pharmacogenomic loci simultaneously, and guidelines for implementation of these data are available from organizations such as the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG). NGS and related technologies are increasing knowledge in the research sphere, yet rates of genomic literacy remain low, resulting in a widening gap in knowledge translation to the patient. Multidisciplinary teams—including physicians, nurses, genetic counsellors, and pharmacists—will need to combine their expertise to deliver optimal pharmacogenomically-informed care.
Collapse
Affiliation(s)
- Catriona Hippman
- Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 2A1, Canada.
- BC Mental Health and Addictions Research Institute, 3rd Floor - 938 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Corey Nislow
- Faculty of Pharmaceutical Sciences, University of British Columbia, 6619-2405 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
75
|
Alshabeeb MA, Deneer VHM, Khan A, Asselbergs FW. Use of Pharmacogenetic Drugs by the Dutch Population. Front Genet 2019; 10:567. [PMID: 31312209 PMCID: PMC6614185 DOI: 10.3389/fgene.2019.00567] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/29/2019] [Indexed: 12/27/2022] Open
Abstract
Introduction The Dutch Pharmacogenetics Working Group (DPWG) indicated a list of actionable genotypes that affect patients’ response to more 50 drugs; these drugs which show variable effects based on patients’ genetic traits were named as pharmacogenetics (PGX) drugs. Preemptive genetic testing before using these drugs may protect certain patients from serious adverse reactions and could help in avoiding treatment failures. The objectives of this study include identifying the rate of PGX drug usage among Dutch population, estimating the level of users who carry the actionable genotypes and determining the main genes involved in drug’s effect variability. Methods Usage of PGX drugs over 2011–2017 by the insured population (an average of 11.4 million) in outpatient clinics in Netherlands was obtained from the publically available GIP databank. The data of 45 drugs were analyzed and their interactions with selected pharmacogenes were estimated. Frequency of actionable genotypes of 249 Dutch parents was obtained from the public database: Genome of Netherlands (GoNL), to identify the pattern of genetic characteristics of Dutch population. Results Over a 7 year period, 51.3 million exposures of patients to PGX drugs were reported with an average of 5.3 exposures per each drug user. One quarterof the exposures (12.4 million) are predicted to be experienced by individuals with actionable genotypes (risky exposures). Up to 60% of the risky exposures (around 7.5 million) were related to drugs metabolized by CYP2D6. SLCO1B1, and CYP2C19 were also identified among the top genes affecting response of drugs users (involved in about 22 and 12.4% of the risky exposures, respectively). Cardiovascular medications were the top prescribed PGX drug class (43%), followed by gastroenterology (29%) and psychiatry/neurology medications (15%). Women use more PGX drugs than men (55.8 vs. 44.2%, respectively) with the majority (84%) of users in both sexes are above 45 years. Conclusion PGX drugs are commonly used in Netherlands. Preemptive panel testing for CYP2D6, SLCO1B1, and CYP2C19 only could be useful to predict 95% of vulnerable patients’ exposures to PGX drugs. Future studies to assess the economic impact of preemptive panel testing on patients of older age are suggested.
Collapse
Affiliation(s)
- Mohammad A Alshabeeb
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Vera H M Deneer
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, Netherlands
| | - Amjad Khan
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Folkert W Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Faculty of Population Health Sciences, Institute of Cardiovascular Science, University College London, London, United Kingdom.,Health Data Research UK and Institute of Health Informatics, University College London, London, United Kingdom
| |
Collapse
|
76
|
Zebrowski AM, Ellis DE, Barg FK, Sperber NR, Bernhardt BA, Denny JC, Dexter PR, Ginsburg GS, Horowitz CR, Johnson JA, Levy MA, Orlando LA, Pollin TI, Skaar TC, Kimmel SE. Qualitative study of system-level factors related to genomic implementation. Genet Med 2019; 21:1534-1540. [PMID: 30467402 PMCID: PMC6533158 DOI: 10.1038/s41436-018-0378-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/09/2018] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Research on genomic medicine integration has focused on applications at the individual level, with less attention paid to implementation within clinical settings. Therefore, we conducted a qualitative study using the Consolidated Framework for Implementation Research (CFIR) to identify system-level factors that played a role in implementation of genomic medicine within Implementing GeNomics In PracTicE (IGNITE) Network projects. METHODS Up to four study personnel, including principal investigators and study coordinators from each of six IGNITE projects, were interviewed using a semistructured interview guide that asked interviewees to describe study site(s), progress at each site, and factors facilitating or impeding project implementation. Interviews were coded following CFIR inner-setting constructs. RESULTS Key barriers included (1) limitations in integrating genomic data and clinical decision support tools into electronic health records, (2) physician reluctance toward genomic research participation and clinical implementation due to a limited evidence base, (3) inadequate reimbursement for genomic medicine, (4) communication among and between investigators and clinicians, and (5) lack of clinical and leadership engagement. CONCLUSION Implementation of genomic medicine is hindered by several system-level barriers to both research and practice. Addressing these barriers may serve as important facilitators for studying and implementing genomics in practice.
Collapse
Affiliation(s)
- Alexis M Zebrowski
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, USA
| | - Darcy E Ellis
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Frances K Barg
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Family Medicine and Community Health, University of Pennsylvania, Philadelphia, PA, USA
| | - Nina R Sperber
- Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Barbara A Bernhardt
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of General Internal Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Paul R Dexter
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Geoffrey S Ginsburg
- Duke Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC, USA
| | - Carol R Horowitz
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Mia A Levy
- Departments of Biomedical Informatics and Medicine, Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lori A Orlando
- Duke Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC, USA
- Division of General Internal Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Toni I Pollin
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Todd C Skaar
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen E Kimmel
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA.
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Medicine, Pearlman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
77
|
Crisafulli C, Romeo PD, Calabrò M, Epasto LM, Alberti S. Pharmacogenetic and pharmacogenomic discovery strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:225-241. [PMID: 35582724 PMCID: PMC8992635 DOI: 10.20517/cdr.2018.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 11/12/2022]
Abstract
Genetic/genomic profiling at a single-patient level is expected to provide critical information for determining inter-individual drug toxicity and potential efficacy in cancer therapy. A better definition of cancer subtypes at a molecular level, may correspondingly complement such pharmacogenetic and pharmacogenomic approaches, for more effective personalized treatments. Current pharmacogenetic/pharmacogenomic strategies are largely based on the identification of known polymorphisms, thus limiting the discovery of novel or rarer genetic variants. Recent improvements in cost and throughput of next generation sequencing (NGS) are now making whole-genome profiling a plausible alternative for clinical procedures. Beyond classical pharmacogenetic/pharmacogenomic traits for drug metabolism, NGS screening programs of cancer genomes may lead to the identification of novel cancer-driving mutations. These may not only constitute novel therapeutic targets, but also effector determinants for metabolic pathways linked to drug metabolism. An additional advantage is that cancer NGS profiling is now leading to discovering targetable mutations, e.g., in glioblastomas and pancreatic cancers, which were originally discovered in other tumor types, thus allowing for effective repurposing of active drugs already on the market.
Collapse
Affiliation(s)
- Concetta Crisafulli
- Department of Biomedical Sciences - BIOMORF, University of Messina, via Consolare Valeria, 98125 Messina, Italy
| | | | - Marco Calabrò
- Department of Biomedical Sciences - BIOMORF, University of Messina, via Consolare Valeria, 98125 Messina, Italy
| | - Ludovica Martina Epasto
- Unit of Medical Genetics, University of Messina, via Consolare Valeria, 98125 Messina, Italy
| | - Saverio Alberti
- Department of Biomedical Sciences - BIOMORF, University of Messina, via Consolare Valeria, 98125 Messina, Italy.,Unit of Medical Genetics, University of Messina, via Consolare Valeria, 98125 Messina, Italy.,Correspondence Address: Prof. Saverio Alberti, Unit of Medical Genetics, BIOMORF Department of Biomedical Sciences, University of Messina, via Consolare Valeria, 98125 Messina, Italy. E-mail:
| |
Collapse
|
78
|
Lee CR, Sriramoju VB, Cervantes A, Howell LA, Varunok N, Madan S, Hamrick K, Polasek MJ, Lee JA, Clarke M, Cicci JD, Weck KE, Stouffer GA. Clinical Outcomes and Sustainability of Using CYP2C19 Genotype-Guided Antiplatelet Therapy After Percutaneous Coronary Intervention. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002069. [PMID: 29615454 DOI: 10.1161/circgen.117.002069] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/05/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND CYP2C19 loss-of-function (LOF) alleles impair clopidogrel effectiveness after percutaneous coronary intervention. The feasibility, sustainability, and clinical impact of using CYP2C19 genotype-guided dual antiplatelet therapy (DAPT) selection in practice remains unclear. METHODS A single-center observational study was conducted in 1193 patients who underwent percutaneous coronary intervention and received DAPT after implementation of an algorithm that recommends CYP2C19 testing in high-risk patients and alternative DAPT (prasugrel or ticagrelor) in LOF allele carriers. The frequency of genotype testing and alternative DAPT selection were the primary implementation end points. Risk of major adverse cardiovascular or cerebrovascular and clinically significant bleeding events over 12 months were compared across genotype and DAPT groups by proportional hazards regression. RESULTS CYP2C19 genotype was obtained in 868 (72.8%) patients. Alternative DAPT was prescribed in 186 (70.7%) LOF allele carriers. CYP2C19 testing (P<0.001) and alternative DAPT use in LOF allele carriers (P=0.001) varied over time. Risk for major adverse cardiovascular or cerebrovascular was significantly higher in LOF carriers prescribed clopidogrel versus alternative DAPT (adjusted hazard ratio, 4.65; 95% confidence interval, 2.22-10.0; P<0.001), whereas no significant difference was observed in those without a LOF allele (adjusted hazard ratio, 1.37; 95% confidence interval, 0.72-2.85; P=0.347). Bleeding event rates were similar across groups (log-rank P=0.816). CONCLUSIONS Implementing CYP2C19 genotype-guided DAPT is feasible and sustainable in a real-world setting but challenging to maintain at a consistently high level of fidelity. The higher risk of major adverse cardiovascular or cerebrovascular associated with clopidogrel use in CYP2C19 LOF allele carriers suggests that use of genotype-guided DAPT in practice may improve clinical outcomes.
Collapse
Affiliation(s)
- Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill.
| | - Vindhya B Sriramoju
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Alexandra Cervantes
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Lucius A Howell
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Nicholas Varunok
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Shivanshu Madan
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Kasey Hamrick
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Melissa J Polasek
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - John Andrew Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Megan Clarke
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Jonathan D Cicci
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - Karen E Weck
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| | - George A Stouffer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy (C.R.L., A.C., K.H., M.J.P., J.A.L.), UNC Center for Pharmacogenomics and Individualized Therapy (C.R.L., K.E.W.), UNC McAllister Heart Institute (C.R.L., G.A.S.), Division of Cardiology, UNC School of Medicine (V.B.S., L.A.H., N.V., S.M., G.A.S.), Department of Pharmacy, UNC HealthCare Medical Center (M.C., J.D.C.), and Department of Pathology and Laboratory Medicine, UNC School of Medicine (K.E.W.), University of North Carolina at Chapel Hill
| |
Collapse
|
79
|
Lin B, Chung WK. Cases in Precision Medicine: The Role of Pharmacogenetics in Precision Prescribing. Ann Intern Med 2019; 170:796-804. [PMID: 31108507 PMCID: PMC7458588 DOI: 10.7326/m18-2357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pharmacogenetics may help physicians deliver individualized treatments based on how a person's genes affect a drug's effects and metabolism. This information can help prevent adverse events or improve drug efficacy by enabling the physician to optimize dosage or to avoid a medication with adverse reactions and to prescribe an alternative therapy. This article discusses the current clinical utility of pharmacogenetic testing in the context of a patient who requires anticoagulation with warfarin.
Collapse
Affiliation(s)
- Bohan Lin
- Columbia University, New York, New York (B.L., W.K.C.)
| | - Wendy K Chung
- Columbia University, New York, New York (B.L., W.K.C.)
| |
Collapse
|
80
|
Afsar NA, Bruckmueller H, Werk AN, Nisar MK, Ahmad HR, Cascorbi I. Implications of genetic variation of common Drug Metabolizing Enzymes and ABC Transporters among the Pakistani Population. Sci Rep 2019; 9:7323. [PMID: 31086207 PMCID: PMC6514210 DOI: 10.1038/s41598-019-43736-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/10/2019] [Indexed: 01/09/2023] Open
Abstract
Genetic polymorphism of drug metabolizing enzymes and transporters may influence drug response. The frequency varies substantially between ethnicities thus having implications on appropriate selection and dosage of various drugs in different populations. The distribution of genetic polymorphisms in healthy Pakistanis has so far not been described. In this study, 155 healthy adults (98 females) were included from all districts of Karachi. DNA was extracted from saliva and genotyped for relevant SNVs in CYP1A1, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 as well as ALDH3A1, GSTA1, ABCB1 and ABCC2. About 64% of the participants were born to parents who were unrelated to each other. There was generally a higher prevalence (p < 0.05) of variant alleles of CYP450 1A2, 2B6, 2C19, 3A5, ALDH3A1, GSTM1 as well as ABCB1 and ABCC2 in this study cohort than in other ethnicities reported in the HapMap database. In contrast, the prevalence of variant alleles was lower in GSTA1. Therefore, in the Pakistani population sample from Karachi a significantly different prevalence of variant drug metabolizing enzymes and ABC transporters was observed as compared to other ethnicities, which could have putative clinical consequences on drug efficacy and safety.
Collapse
Affiliation(s)
- Nasir Ali Afsar
- Jinnah Medical and Dental College, Sohail University, 22-23 Shaheed-e-Millat Road, Karachi, 75400, Pakistan.
| | - Henrike Bruckmueller
- Institute of Experimental and Clinical Pharmacology, Christian Albrechts University Kiel, Hospitalstr. 4, Kiel, 24105, Germany
| | - Anneke Nina Werk
- Institute of Experimental and Clinical Pharmacology, Christian Albrechts University Kiel, Hospitalstr. 4, Kiel, 24105, Germany.,Department of Internal Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Muhammad Kashif Nisar
- Jinnah Medical and Dental College, Sohail University, 22-23 Shaheed-e-Millat Road, Karachi, 75400, Pakistan.,Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - H R Ahmad
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan.,Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, Christian Albrechts University Kiel, Hospitalstr. 4, Kiel, 24105, Germany
| |
Collapse
|
81
|
Shotelersuk V, Tongsima S, Pithukpakorn M, Eu‐ahsunthornwattana J, Mahasirimongkol S. Precision medicine in Thailand. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2019; 181:245-253. [DOI: 10.1002/ajmg.c.31694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of MedicineChulalongkorn University Bangkok Thailand
- Excellence Center for Medical GeneticsKing Chulalongkorn Memorial Hospital, the Thai Red Cross Society Bangkok Thailand
| | - Sissades Tongsima
- National Center for Genetic Engineering and BiotechnologyNational Science and Technology Development Agency Pathum Thani Thailand
| | - Manop Pithukpakorn
- Division of Medical Genetics, Department of MedicineFaculty of Medicine Siriraj Hospital, Mahidol University Bangkok Thailand
- Siriraj Center of Research Excellence in Precision MedicineFaculty of Medicine Siriraj Hospital, Mahidol University Bangkok Thailand
| | - Jakris Eu‐ahsunthornwattana
- Department of Community MedicineFaculty of Medicine Ramathibodi Hospital, Mahidol University Bangkok Thailand
- Division of Medical Genetics and Molecular Medicine, Department of Internal Medicine, Faculty of Medicine Ramathibodi HospitalMahidol University Bangkok Thailand
| | - Surakameth Mahasirimongkol
- Medical Genetics Center, Medical Life Sciences Institute, Department of Medical SciencesMinistry of Public Health Nonthaburi Thailand
| |
Collapse
|
82
|
Daneshjou R, Huddart R, Klein TE, Altman RB. Pharmacogenomics in dermatology: tools for understanding gene-drug associations. ACTA ACUST UNITED AC 2019; 38:E19-E24. [PMID: 31051019 DOI: 10.12788/j.sder.2019.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Pharmacogenomics aims to associate human genetic variability with differences in drug phenotypes in order to tailor drug treatment to individual patients. The massive amount of genetic data generated from large cohorts of patients with variable drug phenotypes have led to advances in this field. Understanding the application of pharmacogenomics in dermatology could inform clinical practice and provide insight for future research. The Pharmacogenomics Knowledge Base and the Clinical Pharmacogenetics Implementation Consortium are among the resources to help clinicians and researchers navigate the many gene-drug associations that have already been discovered. The implementation of clinical pharmacogenomics within health care systems remains an area of ongoing development. This review provides an introduction to the field of pharmacogenomics and to current pharmacogenomics resources using examples of gene-drug associations relevant to the field of dermatology.
Collapse
Affiliation(s)
- Roxana Daneshjou
- Department of Dermatology, Stanford School of Medicine, Redwood City, California.
| | - Rachel Huddart
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, California
| | - Teri E Klein
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, California.,Department of Medicine, Stanford School of Medicine, Stanford, California
| | - Russ B Altman
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, California.,Department of Medicine, Stanford School of Medicine, Stanford, California.,Department of Biomedical Engineering, Stanford Schools of Engineering & Medicine, Stanford, California.,Department of Genetics, Stanford School of Medicine, Stanford, California
| |
Collapse
|
83
|
Abstract
Massively parallel sequencing is emerging from research settings into clinical practice, helping the vision of precision medicine to become a reality. The most successful applications are using the tools of implementation science within the framework of the learning health-care system. This article examines the application of massively parallel sequencing to four clinical scenarios: pharmacogenomics, diagnostic testing, somatic testing for molecular tumor characterization, and population screening. For each application, it highlights an exemplar program to illustrate the enablers and challenges of implementation. International examples are also presented. These early lessons will allow other programs to account for these factors, helping to accelerate the implementation of precision medicine and health.
Collapse
Affiliation(s)
- Marc S Williams
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania 17822-2620, USA;
| |
Collapse
|
84
|
Vassy JL, Stone A, Callaghan JT, Mendes M, Meyer LJ, Pratt VM, Przygodzki RM, Scheuner MT, Wang-Rodriguez J, Schichman SA. Pharmacogenetic testing in the Veterans Health Administration (VHA): policy recommendations from the VHA Clinical Pharmacogenetics Subcommittee. Genet Med 2019; 21:382-390. [PMID: 29858578 PMCID: PMC6274593 DOI: 10.1038/s41436-018-0057-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/26/2018] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The Veterans Health Administration (VHA) Clinical Pharmacogenetics Subcommittee is charged with making recommendations about whether specific pharmacogenetic tests should be used in healthcare at VHA facilities. We describe a process to inform VHA pharmacogenetic testing policy. METHODS After developing consensus definitions of clinical validity and utility, the Subcommittee identified salient drug-gene pairs with potential clinical application in VHA. Members met monthly to discuss each drug-gene pair, the evidence of clinical utility for the associated pharmacogenetic test, and any VHA-specific testing considerations. The Subcommittee classified each test as strongly recommended, recommended, or not routinely recommended before drug initiation. RESULTS Of 30 drug-gene pair tests reviewed, the Subcommittee classified 4 (13%) as strongly recommended, including HLA-B*15:02 for carbamazepine-associated Stevens-Johnston syndrome and G6PD for rasburicase-associated hemolytic anemia; 12 (40%) as recommended, including CYP2D6 for codeine toxicity; and 14 (47%) as not routinely recommended, such as CYP2C19 for clopidogrel dosing. CONCLUSION Only half of drug-gene pairs with high clinical validity received Subcommittee support for policy promoting their widespread use across VHA. The Subcommittee generally found insufficient evidence of clinical utility or available, effective alternative strategies for the remainders. Continual evidence review and rigorous outcomes research will help promote the translation of pharmacogenetic discovery to healthcare.
Collapse
Affiliation(s)
- Jason L Vassy
- Section of General Internal Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - John T Callaghan
- Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Laurence J Meyer
- Office of Specialty Care Services, Veterans Health Administration, Washington, DC, USA
| | - Victoria M Pratt
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ronald M Przygodzki
- Office of Research & Development, US Department of Veterans Affairs, Washington, DC, USA
| | - Maren T Scheuner
- Division of Medical Genetics, Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Jessica Wang-Rodriguez
- VA San Diego Healthcare System, San Diego, California, USA
- University of California San Diego, San Diego, California, USA
| | - Steven A Schichman
- Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
85
|
Haga SB, Kantor A. Horizon Scan Of Clinical Laboratories Offering Pharmacogenetic Testing. Health Aff (Millwood) 2019; 37:717-723. [PMID: 29733708 DOI: 10.1377/hlthaff.2017.1564] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pharmacogenetic (PGx) testing involves the analysis of genes known to affect response to medications. The field has been projected as a leading application of personalized or precision medicine, but the use of PGx tests has been stymied, in part, by the lack of clinical evidence of utility and reported low provider awareness. Another factor is the availability of testing. The range and types of PGx tests available have not been assessed to date. In the period September 2017-January 2018 we analyzed the numbers and types of PGx tests offered by clinical testing laboratories in the US. Of the 111 such labs that we identified, we confirmed that 76 offered PGx testing services. Of these, 31 offered only tests for single genes; 30 offered only tests for multiple genes; and 15 offered both types of tests. Collectively, 45 laboratories offered 114 multigene panel tests covering 295 genes. The majority of these tests did not have any clinical guidelines. PGx tests vary in type and makeup, which presents challenges in appropriate test evaluation and selection for providers, insurers, health systems, and patients alike.
Collapse
Affiliation(s)
- Susanne B Haga
- Susanne B. Haga ( ) is an associate professor of medicine at the Duke University School of Medicine, in Durham, North Carolina
| | - Ariel Kantor
- Ariel Kantor is an undergraduate research assistant at Duke University
| |
Collapse
|
86
|
Vassy JL, Brunette CA, Majahalme N, Advani S, MacMullen L, Hau C, Zimolzak AJ, Miller SJ. The Integrating Pharmacogenetics in Clinical Care (I-PICC) Study: Protocol for a point-of-care randomized controlled trial of statin pharmacogenetics in primary care. Contemp Clin Trials 2018; 75:40-50. [PMID: 30367991 PMCID: PMC8119226 DOI: 10.1016/j.cct.2018.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/04/2018] [Accepted: 10/16/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND The association between the SLCO1B1 rs4149056 variant and statin-associated muscle symptoms (SAMS) is well validated, but the clinical utility of its implementation in patient care is unknown. DESIGN The Integrating Pharmacogenetics in Clinical Care (I-PICC) Study is a pseudo-cluster randomized controlled trial of SLCO1B1 genotyping among statin-naïve primary care and women's health patients across the Veteran Affairs Boston Healthcare System. Eligible patients of enrolled primary care providers are aged 40-75 and have elevated risk of cardiovascular disease by American College of Cardiology/American Heart Association (ACC/AHA) guidelines. Patients give consent by telephone in advance of an upcoming appointment, but they are enrolled only if and when their provider co-signs an order for SLCO1B1 testing, performed on a blood sample already collected in clinical care. Enrolled patients are randomly allocated to have their providers receive results through the electronic health record at baseline (PGx + arm) versus after 12 months (PGx- arm). The primary outcome is the change in low-density lipoprotein cholesterol (LDL-C) after one year. Secondary outcomes are concordance with Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for simvastatin prescribing, concordance with ACC/AHA guidelines for statin use, and incidence of SAMS. With 408 patients, the study has >80% power to exclude a between-group LDL-C difference of 10 mg/dL (non-inferiority design) and to detect between-group differences of 15% in CPIC guideline concordance (superiority design). CONCLUSION The outcomes of the I-PICC Study will inform the clinical utility of preemptive SLCO1B1 testing in the routine practice of medicine, including its proposed benefits and unforeseen risks.
Collapse
Affiliation(s)
- Jason L Vassy
- VA Boston Healthcare System, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, MA, USA.
| | | | | | | | | | - Cynthia Hau
- VA Boston Healthcare System, Boston, MA, USA
| | - Andrew J Zimolzak
- VA Boston Healthcare System, Boston, MA, USA; Boston University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
87
|
Chambliss AB, Marzinke MA. Clinical Pharmacogenetics for Precision Medicine: Successes and Setbacks. J Appl Lab Med 2018; 3:474-486. [PMID: 33636912 DOI: 10.1373/jalm.2017.023127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/05/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND Pharmacogenetics is a key component in the delivery of therapeutics to maximize pharmacologic efficacy and minimize toxicity. There are numerous identified gene-drug pairs that demonstrate the utility of pharmacogenetics testing for drug or dose selection. Although some of these pairs have translated into clinical use, pharmacogenetic testing has not yet made its way into routine clinical practice at many institutions. CONTENT This review provides an overview of clinically actionable pharmacogenetics in precision medicine. Examples of successfully implemented gene-drug pairs, along with common testing methodologies and guidelines for application, are discussed. Remaining barriers to widespread clinical implementation are also examined. SUMMARY There is a recognized role for genotyping in the guidance of therapeutic drug regimens and the prevention of adverse drug reactions. Evidence-based guidelines are available to aid in the selection of treatment upon pharmacogenetics testing for established gene-drug pairs. Multidisciplinary clinical collaboration and clinical decision support tools will be critical for widespread adoption, and financial reimbursement barriers remain.
Collapse
Affiliation(s)
- Allison B Chambliss
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Mark A Marzinke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
88
|
Frick A, Benton C, Suzuki O, Dong O, Howard R, El-Sabae H, Wiltshire T. Implementing Clinical Pharmacogenomics in the Classroom: Student Pharmacist Impressions of an Educational Intervention Including Personal Genotyping. PHARMACY 2018; 6:pharmacy6040115. [PMID: 30360487 PMCID: PMC6306770 DOI: 10.3390/pharmacy6040115] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 01/07/2023] Open
Abstract
Pharmacogenomics provides a personalized approach to pharmacotherapy by using genetic information to guide drug dosing and selection. However, partly due to lack of education, pharmacogenomic testing has not been fully implemented in clinical practice. With pharmacotherapy training and patient accessibility, pharmacists are ideally suited to apply pharmacogenomics to patient care. Student pharmacists (n = 222) participated in an educational intervention that included voluntary personal genotyping using 23andMe. Of these, 31% of students completed both pre- and post-educational interventions to evaluate their attitudes and confidence towards the use of pharmacogenomics data in clinical decision making, and 55% of this paired subset obtained personal genotyping. McNemar’s test and the Wilcoxon signed-rank test were used to analyze responses. Following the educational intervention, students regardless of genotyping were more likely to recommend personal genotyping (36% post-educational intervention versus 19% pre-educational intervention, p = 0.0032), more confident in using pharmacogenomics in the management of drug therapy (51% post-educational intervention versus 29% pre-educational intervention, p = 0.0045), and more likely to believe that personalized genomics would have an important role in their future pharmacy career (90% post-educational intervention versus 51% pre-educational intervention, p = 0.0072) compared to before receiving the educational intervention. This educational intervention positively influenced students’ attitudes and confidence regarding pharmacogenomics in the clinical setting. Future studies will examine the use of next-generation sequencing assays that selectively examine pharmacogenes in the education of student pharmacists.
Collapse
Affiliation(s)
- Amber Frick
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Cristina Benton
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Oscar Suzuki
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Olivia Dong
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Rachel Howard
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Hijrah El-Sabae
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Tim Wiltshire
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
89
|
Vassy JL, Chun S, Advani S, Ludin SA, Smith JG, Alligood EC. Impact of SLCO1B1 Pharmacogenetic Testing on Patient and Healthcare Outcomes: A Systematic Review. Clin Pharmacol Ther 2018; 106:360-373. [PMID: 30137643 DOI: 10.1002/cpt.1223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/14/2018] [Indexed: 12/22/2022]
Abstract
Demonstrated improvements in patient outcomes will facilitate the clinical implementation of pharmacogenetic testing. Using the association between solute carrier organic anion transporter family member 1B1 (SLCO1B1) and statin-associated muscle symptoms (SAMSs) as a model, we conducted a systematic review of patient outcomes after delivery of SLCO1B1 results. Using PubMed and Embase searches through December 19, 2017, we identified 37 eligible records reporting preliminary or final outcomes, including six studies delivering only SLCO1B1 results and five large healthcare system-based implementation projects of multipharmacogene panels. Two small trials have demonstrated at least short-term improvements in low-density lipoprotein cholesterol after SLCO1B1 testing among previously statin intolerant patients. Evidence from large implementation projects suggests that SLCO1B1 results may change prescribing patterns for some high-risk patients. No study has reported improvements in SAMSs or cardiovascular events or tracked the economic outcomes of SLCO1B1 testing. Ongoing studies should collect and report outcomes relevant to pharmacogenetics stakeholders.
Collapse
Affiliation(s)
- Jason L Vassy
- Veterans Affairs (VA) VA Boston Healthcare System, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sojeong Chun
- Veterans Affairs (VA) VA Boston Healthcare System, Boston, Massachusetts, USA.,Massachusetts College of Pharmacy and Health Sciences University, Boston, Massachusetts, USA
| | - Sanjay Advani
- Veterans Affairs (VA) VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Sophie A Ludin
- Veterans Affairs (VA) VA Boston Healthcare System, Boston, Massachusetts, USA.,Cornell University, Ithaca, New York, USA
| | - Jason G Smith
- Veterans Affairs (VA) VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Elaine C Alligood
- Veterans Affairs (VA) VA Boston Healthcare System, Boston, Massachusetts, USA
| |
Collapse
|
90
|
Abstract
Pharmacogenetics, a major component of individualized or precision medicine, relies on human genetic diversity. The remarkable developments in sequencing technologies have revealed that the number of genetic variants modulating drug action is much higher than previously thought and that a true personalized prediction of drug response requires attention to rare mutations (minor allele frequency, MAF<1%) in addition to polymorphisms (MAF>1%) in pharmacogenes. This has major implications for the conceptual development and clinical implementation of pharmacogenetics. Drugs used in cancer treatment have been major targets of pharmacogenetics studies, encompassing both germline polymorphisms and somatic variants in the tumor genome. The present overview, however, has a narrower scope and is focused on germline cancer pharmacogenetics, more specifically, on drug/gene pairs for which pharmacogenetics-informed prescription guidelines have been published by the Clinical Pharmacogenetics Implementation Consortium and/or the Dutch Pharmacogenetic Working Group, namely, thiopurines/TPMT, fluoropyrimidines/UGT1A1, irinotecan/UGT1A1 and tamoxifen/CYP2D6. I begin by reviewing the general principles of pharmacogenetics-informed prescription, pharmacogenetics testing and the perceived barriers to the adoption of routine pharmacogenetics testing in clinical practice. Then, I highlight aspects of the pharmacogenetics testing of the selected drug-gene pairs and finally present pharmacogenetics data from Brazilian studies pertinent to these drug-gene pairs. I conclude with the notion that pharmacogenetics testing has the potential to greatly benefit patients by enabling precision medicine applied to drug therapy, ensuring better efficacy and reducing the risk of adverse effects.
Collapse
Affiliation(s)
- Guilherme Suarez-Kurtz
- Instituto Nacional de Cancer, Rio de Janeiro, RJ, BR
- Rede Nacional de Farmacogenetica, Rio de Janeiro, RJ, BR
- *Corresponding author. E-mail:
| |
Collapse
|
91
|
Luzum JA, Cheung JC. Does cardiology hold pharmacogenetics to an inconsistent standard? A comparison of evidence among recommendations. Pharmacogenomics 2018; 19:1203-1216. [PMID: 30196751 PMCID: PMC6219446 DOI: 10.2217/pgs-2018-0097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022] Open
Abstract
Current guideline recommendations for pharmacogenetic testing for clopidogrel by the American Heart Association/American College of Cardiology (AHA/ACC) contradict the Clinical Pharmacogenetics Implementation Consortium and the US FDA. The AHA/ACC recommends against routine pharmacogenetic testing for clopidogrel because no randomized controlled trials have demonstrated that testing improves patients' outcomes. However the AHA/ACC and the National Comprehensive Cancer Network (NCCN) recommend other pharmacogenetic tests in the absence of randomized controlled trials evidence. Using clopidogrel as a case example, we compared the evidence for other pharmacogenetic tests recommended by the AHA/ACC and NCCN. In patients that received percutaneous coronary intervention, the evidence supporting pharmacogenetic testing for clopidogrel is stronger than other pharmacogenetic tests recommended by the AHA/ACC and NCCN.
Collapse
Affiliation(s)
- Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Jason C Cheung
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
- Department of Pharmacy, Baptist Health Floyd, New Albany, IN, USA
| |
Collapse
|
92
|
Mills R, Haga SB. Qualitative user evaluation of a revised pharmacogenetic educational toolkit. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2018; 11:139-146. [PMID: 30214267 PMCID: PMC6128278 DOI: 10.2147/pgpm.s169648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Introduction Pharmacogenetic (PGx) testing is a leading application for personalized and precision medicine; however, there are barriers, including limited provider and patient understanding, which affect its uptake. There is a need for tools that can enhance the patient and provider experience with testing and promoting the shared and informed decision-making. Materials and methods In this study, we sought to gather additional feedback on a PGx toolkit comprised of four educational tools that had been previously evaluated through an online survey by pharmacists. Specifically, we conducted semi-structured interviews with pharmacists and members of the public regarding their understanding and utility of the toolkit and its individual components. Results Participants found three of the four toolkit components, a test information sheet, flipbook, and results sheet, to be useful and important. The fourth component, results card, was viewed less favorably. Participants differed in their preference for medical jargon and detailed results nomenclature (namely star * alleles). Conclusion User input during the development of educational materials is essential for optimizing utilization, effectiveness, and comprehension.
Collapse
Affiliation(s)
- Rachel Mills
- Duke Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC, USA,
| | - Susanne B Haga
- Duke Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, NC, USA,
| |
Collapse
|
93
|
Klein MD, Lee CR, Stouffer GA. Clinical outcomes of CYP2C19 genotype-guided antiplatelet therapy: existing evidence and future directions. Pharmacogenomics 2018; 19:1039-1046. [DOI: 10.2217/pgs-2018-0072] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
It is well established that the CYP2C19 nonfunctional *2 and *3 polymorphisms impair the bioactivation and antiplatelet effects of clopidogrel, and increase the risk of adverse cardiovascular events following percutaneous coronary intervention. In contrast, CYP2C19 genotype does not impact clinical response to prasugrel or ticagrelor. Recent studies have evaluated the impact of CYP2C19 genotype-guided selection of antiplatelet therapy on clinical outcomes and begun to close some of the gaps in knowledge and uncertainty that have impeded widespread clinical implementation of this precision medicine approach. This review will critically evaluate recent data and offer new insight into the potential clinical utility of genotype-guided antiplatelet therapy in the context of current clinical practice guidelines.
Collapse
Affiliation(s)
- Melissa D Klein
- Division of Cardiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Craig R Lee
- McAllister Heart Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Pharmacotherapy & Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Pharmacogenomics & Individualized Therapy, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - George A Stouffer
- Division of Cardiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
94
|
Hertz DL, Glatz A, Pasternak AL, Lonigro RJ, Vats P, Wu YM, Anderson B, Rabban E, Mora E, Frank K, Robinson DR, Mody RJ, Chinnaiyan A. Integration of Germline Pharmacogenetics Into a Tumor Sequencing Program. JCO Precis Oncol 2018; 2:PO.18.00011. [PMID: 32832831 PMCID: PMC7434089 DOI: 10.1200/po.18.00011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Evidence-based guidelines inform treatment decisions for patients for whom germline genetic information is available. Our real-time tumor sequencing program, which makes precision treatment decisions for patients with cancer, produces matched germline information, providing a unique opportunity to efficiently implement pharmacogenetics and benefit patients. METHODS The germline genetic database from the Michigan Oncology Sequencing (MI-Oncoseq) program was searched for 21 clinically actionable polymorphisms in five cancer-relevant genes: TPMT, DPYD, CYP2C19, CYP3A5, and UGT1A1. Residual germ line DNA was sent to an external Clinical Laboratory Improvement Amendments-approved laboratory for confirmatory genotyping. The medical records of MI-Oncoseq patients with actionable phenotypes were searched for receipt of relevant drugs and to determine whether having genetic information at the time of treatment would have led to a treatment recommendation. RESULTS All nine variants in TPMT, DPYD, and CYP2C19 that were detected in MI-Oncoseq were confirmed by external genotyping. Genotype determinations could not be made for CYP3A5*3, UGT1A1*28, or UGT1A1*80. On the basis of retrospective assessment of 115 adult and pediatric patient records, 4.3% (n = 5) had a potentially clinically actionable phenotype for TPMT, DPYD, or CYP2C19 and received a relevant medication. After accounting for differences in adult and pediatric recommendations, three of these patients could have received a treatment recommendation at the time of prescribing. CONCLUSION Germline genotype determinations for TPMT, DPYD, and CYP2C19 can be used to make evidence-based treatment recommendations in MI-Oncoseq patients. Although the proportion of patients for whom recommendations can be made is small, this added value to MI-Oncoseq and patient care comes at no additional genotyping cost. Pharmacogenetic assessment should be integrated into tumor sequencing programs that genotype matched germline DNA; however, the complexity and additional cost of implementing pharmacogenetics remain challenging.
Collapse
Affiliation(s)
- Daniel L. Hertz
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Andrew Glatz
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Amy L. Pasternak
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Robert J. Lonigro
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Pankaj Vats
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Yi-Mi Wu
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Bailey Anderson
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Erica Rabban
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Erika Mora
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Kevin Frank
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Dan R. Robinson
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Rajen J. Mody
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| | - Arul Chinnaiyan
- Daniel L. Hertz, Andrew Glatz, Amy L. Pasternak, and Erika Mora, University of Michigan College of Pharmacy; Robert J. Lonigro, Pankaj Vats, Yi-Mi Wu, Bailey Anderson, Erica Rabban, Kevin Frank, Dan R. Robinson, Rajen J. Mody, and Arul Chinnaiyan, Michigan Medicine, Ann Arbor, MI
| |
Collapse
|
95
|
Barbarino JM, Whirl‐Carrillo M, Altman RB, Klein TE. PharmGKB: A worldwide resource for pharmacogenomic information. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1417. [PMID: 29474005 PMCID: PMC6002921 DOI: 10.1002/wsbm.1417] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/04/2023]
Abstract
As precision medicine becomes increasingly relevant in healthcare, the field of pharmacogenomics (PGx) also continues to gain prominence in the clinical setting. Leading institutions have begun to implement PGx testing and the amount of published PGx literature increases yearly. The Pharmacogenomics Knowledgebase (PharmGKB; www.pharmgkb.org) is one of the foremost worldwide resources for PGx knowledge, and the organization has been adapting and refocusing its mission along with the current revolution in genomic medicine. The PharmGKB website provides a diverse array of PGx information, from annotations of the primary literature to guidelines for adjusting drug treatment based on genetic information. It is freely available and accessible to everyone from researchers to clinicians to everyday citizens. PharmGKB was found over 17 years ago, but continues to be a vital resource for the entire PGx community and the general public. This article is categorized under: Translational, Genomic, and Systems Medicine > Translational Medicine.
Collapse
Affiliation(s)
- Julia M. Barbarino
- Department of Biomedical Data SciencesStanford UniversityStanfordCalifornia
| | | | - Russ B. Altman
- Department of Biomedical EngineeringStanford UniversityStanfordCalifornia
- Department of GeneticsStanford UniversityStanfordCalifornia
| | - Teri E. Klein
- Department of Biomedical Data SciencesStanford UniversityStanfordCalifornia
- Department of MedicineStanford UniversityStanfordCalifornia
| |
Collapse
|
96
|
Dong OM, Li A, Suzuki O, Oni-Orisan A, Gonzalez R, Stouffer GA, Lee CR, Wiltshire T. Projected impact of a multigene pharmacogenetic test to optimize medication prescribing in cardiovascular patients. Pharmacogenomics 2018; 19:771-782. [PMID: 29793377 PMCID: PMC6367721 DOI: 10.2217/pgs-2018-0049] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM To determine the projected impact of a multigene pharmacogenetic (PGx) test on medication prescribing. MATERIALS & METHODS A retrospective analysis was conducted with 122 cardiac catheterization laboratory patients undergoing angiography for eligibility of potential PGx-guided interventions that could have occurred if multigene PGx information was pre-emptively available at the time of the procedure. Medication data and presence of actionable at-risk genotypes were used to determine eligibility of a PGx intervention. RESULTS 20% of the study population (n = 24) would have qualified for at least one PGx-based medication intervention per US FDA or Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines within 6 months of their cardiac catheterization procedure. Commonly encountered gene-drug pairs for these interventions included: CYP2C19 for clopidogrel and antidepressants, CYP2D6 for antidepressants and codeine, SLCO1B1 for simvastatin, and VKORC1/CYP2C9 for warfarin. CONCLUSION Pre-emptive use of a multigene PGx test in the cardiac catheterization laboratory offers potential to reduce adverse medication outcomes.
Collapse
Affiliation(s)
- Olivia M Dong
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amy Li
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Oscar Suzuki
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, UCSF School of Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ricardo Gonzalez
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - George A Stouffer
- UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Division of Cardiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Lee
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tim Wiltshire
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Center for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
97
|
Roden DM, Van Driest SL, Mosley JD, Wells QS, Robinson JR, Denny JC, Peterson JF. Benefit of Preemptive Pharmacogenetic Information on Clinical Outcome. Clin Pharmacol Ther 2018; 103:787-794. [PMID: 29377064 PMCID: PMC6134843 DOI: 10.1002/cpt.1035] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/08/2018] [Accepted: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The development of new knowledge around the genetic determinants of variable drug action has naturally raised the question of how this new knowledge can be used to improve the outcome of drug therapy. Two broad approaches have been taken: a point-of-care approach in which genotyping for specific variant(s) is undertaken at the time of drug prescription, and a preemptive approach in which multiple genetic variants are typed in an individual patient and the information archived for later use when a drug with a "pharmacogenetic story" is prescribed. This review addresses the current state of implementation, the rationale for these approaches, and barriers that must be overcome. Benefits to pharmacogenetic testing are only now being defined and will be discussed.
Collapse
Affiliation(s)
- Dan M. Roden
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN
- Department of Pharmacology, Vanderbilt University Medical Center Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center Nashville, TN
| | - Sara L. Van Driest
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN
- Department of Pediatrics, Vanderbilt University Medical Center Nashville, TN
| | - Jonathan D. Mosley
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center Nashville, TN
| | - Quinn S. Wells
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN
| | - Jamie R. Robinson
- Department of Biomedical Informatics, Vanderbilt University Medical Center Nashville, TN
- Department of Surgery, Vanderbilt University Medical Center Nashville, TN
| | - Joshua C. Denny
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center Nashville, TN
| | - Josh F. Peterson
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center Nashville, TN
| |
Collapse
|
98
|
Mills RA, Eichmeyer JN, Williams LM, Muskett JA, Schmidlen TJ, Maloney KA, Lemke AA. Patient Care Situations Benefiting from Pharmacogenomic Testing. CURRENT GENETIC MEDICINE REPORTS 2018. [DOI: 10.1007/s40142-018-0136-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
99
|
Hintzsche JD, Yoo M, Kim J, Amato CM, Robinson WA, Tan AC. IMPACT web portal: oncology database integrating molecular profiles with actionable therapeutics. BMC Med Genomics 2018; 11:26. [PMID: 29697364 PMCID: PMC5918430 DOI: 10.1186/s12920-018-0350-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background With the advancement of next generation sequencing technology, researchers are now able to identify important variants and structural changes in DNA and RNA in cancer patient samples. With this information, we can now correlate specific variants and/or structural changes with actionable therapeutics known to inhibit these variants. We introduce the creation of the IMPACT Web Portal, a new online resource that connects molecular profiles of tumors to approved drugs, investigational therapeutics and pharmacogenetics associated drugs. Results IMPACT Web Portal contains a total of 776 drugs connected to 1326 target genes and 435 target variants, fusion, and copy number alterations. The online IMPACT Web Portal allows users to search for various genetic alterations and connects them to three levels of actionable therapeutics. The results are categorized into 3 levels: Level 1 contains approved drugs separated into two groups; Level 1A contains approved drugs with variant specific information while Level 1B contains approved drugs with gene level information. Level 2 contains drugs currently in oncology clinical trials. Level 3 provides pharmacogenetic associations between approved drugs and genes. Conclusion IMPACT Web Portal allows for sequencing data to be linked to actionable therapeutics for translational and drug repurposing research. The IMPACT Web Portal online resource allows users to query genes and variants to approved and investigational drugs. We envision that this resource will be a valuable database for personalized medicine and drug repurposing. IMPACT Web Portal is freely available for non-commercial use at http://tanlab.ucdenver.edu/IMPACT.
Collapse
Affiliation(s)
- Jennifer D Hintzsche
- Division of Medical Oncology, Department of Medicine, Translational Bioinformatics and Cancer Systems Biology Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Division of Medical Oncology, Department of Medicine, Robinson Melanoma Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Minjae Yoo
- Division of Medical Oncology, Department of Medicine, Translational Bioinformatics and Cancer Systems Biology Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jihye Kim
- Division of Medical Oncology, Department of Medicine, Translational Bioinformatics and Cancer Systems Biology Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carol M Amato
- Division of Medical Oncology, Department of Medicine, Robinson Melanoma Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - William A Robinson
- Division of Medical Oncology, Department of Medicine, Robinson Melanoma Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aik Choon Tan
- Division of Medical Oncology, Department of Medicine, Translational Bioinformatics and Cancer Systems Biology Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
100
|
Lemke AA, Hulick PJ, Wake DT, Wang C, Sereika AW, Yu KD, Glaser NS, Dunnenberger HM. Patient perspectives following pharmacogenomics results disclosure in an integrated health system. Pharmacogenomics 2018; 19:321-331. [DOI: 10.2217/pgs-2017-0191] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: To assess patient perceptions and utilization of pharmacogenomics (PGx) testing in an integrated community health system. Methods: Fifty-seven patients completed an online survey assessing their experiences with PGx testing offered through two methods: a designated PGx clinic or direct access in-home testing. Results: The majority of participants perceived PGx testing as helpful in their healthcare and reported understanding their results. Some had concerns about privacy and discrimination; most lacked familiarity with the Genetic Information Nondiscrimination Act. There were no significant differences in views between participants tested through either model. Conclusion: Participants reported value in both methods of PGx testing. Patient experiences, understanding and result utilization will play an important role in informing future development and implementation of PGx programs.
Collapse
Affiliation(s)
- Amy A Lemke
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| | - Peter J Hulick
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| | - Dyson T Wake
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| | - Chi Wang
- Biostatistics & Research Informatics, NorthShore University HealthSystem, 1001 University Place, Suite 146, Evanston, IL 60201, USA
| | - Annette W Sereika
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| | - Kristen Dilzell Yu
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| | - Nicole S Glaser
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| | - Henry M Dunnenberger
- Center for Personalized Medicine, NorthShore University HealthSystem, 1001 University Place, Suite 160, Evanston, IL 60201, USA
| |
Collapse
|