1
|
Boyle GE, Sitko KA, Galloway JG, Haddox HK, Bianchi AH, Dixon A, Wheelock MK, Vandi AJ, Wang ZR, Thomson RES, Garge RK, Rettie AE, Rubin AF, Geck RC, Gillam EMJ, DeWitt WS, Matsen FA, Fowler DM. Deep mutational scanning of CYP2C19 in human cells reveals a substrate specificity-abundance tradeoff. Genetics 2024; 228:iyae156. [PMID: 39319420 PMCID: PMC11538415 DOI: 10.1093/genetics/iyae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/31/2024] [Indexed: 09/26/2024] Open
Abstract
The cytochrome P450s enzyme family metabolizes ∼80% of small molecule drugs. Variants in cytochrome P450s can substantially alter drug metabolism, leading to improper dosing and severe adverse drug reactions. Due to low sequence conservation, predicting variant effects across cytochrome P450s is challenging. Even closely related cytochrome P450s like CYP2C9 and CYP2C19, which share 92% amino acid sequence identity, display distinct phenotypic properties. Using variant abundance by massively parallel sequencing, we measured the steady-state protein abundance of 7,660 single amino acid variants in CYP2C19 expressed in cultured human cells. Our findings confirmed critical positions and structural features essential for cytochrome P450 function, and revealed how variants at conserved positions influence abundance. We jointly analyzed 4,670 variants whose abundance was measured in both CYP2C19 and CYP2C9, finding that the homologs have different variant abundances in substrate recognition sites within the hydrophobic core. We also measured the abundance of all single and some multiple wild type amino acid exchanges between CYP2C19 and CYP2C9. While most exchanges had no effect, substitutions in substrate recognition site 4 reduced abundance in CYP2C19. Double and triple mutants showed distinct interactions, highlighting a region that points to differing thermodynamic properties between the 2 homologs. These positions are known contributors to substrate specificity, suggesting an evolutionary tradeoff between stability and enzymatic function. Finally, we analyzed 368 previously unannotated human variants, finding that 43% had decreased abundance. By comparing variant effects between these homologs, we uncovered regions underlying their functional differences, advancing our understanding of this versatile family of enzymes.
Collapse
Affiliation(s)
- Gabriel E Boyle
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Katherine A Sitko
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jared G Galloway
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Hugh K Haddox
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Aisha Haley Bianchi
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Ajeya Dixon
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Melinda K Wheelock
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Allyssa J Vandi
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Ziyu R Wang
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Raine E S Thomson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4067, Australia
| | - Riddhiman K Garge
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
| | - Allan E Rettie
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Alan F Rubin
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Renee C Geck
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth M J Gillam
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4067, Australia
| | - William S DeWitt
- Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Frederick A Matsen
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98109, USA
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2
|
Peña-Martín MC, Marcos-Vadillo E, García-Berrocal B, Heredero-Jung DH, García-Salgado MJ, Lorenzo-Hernández SM, Larrue R, Lenski M, Drevin G, Sanz C, Isidoro-García M. A Comparison of Molecular Techniques for Improving the Methodology in the Laboratory of Pharmacogenetics. Int J Mol Sci 2024; 25:11505. [PMID: 39519058 PMCID: PMC11546559 DOI: 10.3390/ijms252111505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
One of the most critical goals in healthcare is safe and effective drug therapy, which is directly related to an individual's response to treatment. Precision medicine can improve drug safety in many scenarios, including polypharmacy, and it requires the development of new genetic characterization methods. In this report, we use real-time PCR, microarray techniques, and mass spectrometry (MALDI-TOF), which allows us to compare them and identify the potential benefits of technological improvements, leading to better quality medical care. These comparative studies, as part of our pharmacogenetic Five-Step Precision Medicine (5SPM) approach, reveal the superiority of mass spectrometry over the other methods analyzed and highlight the importance of updating the laboratory's pharmacogenetic methodology to identify new variants with clinical impact.
Collapse
Affiliation(s)
- María Celsa Peña-Martín
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Pharmacology-Toxicology and Pharmacovigilance Department, Angers University Hospital, F-49100 Angers, France;
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Elena Marcos-Vadillo
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Belén García-Berrocal
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - David Hansoe Heredero-Jung
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - María Jesús García-Salgado
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Sandra Milagros Lorenzo-Hernández
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
| | - Romain Larrue
- CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, University of Lille, F-59000 Lille, France;
| | - Marie Lenski
- CHU Lille, Institut Pasteur de Lille, ULR 4483, IMPECS-IMPact of the Chemical Environment on Health, University of Lille, F-59000 Lille, France;
| | - Guillaume Drevin
- Pharmacology-Toxicology and Pharmacovigilance Department, Angers University Hospital, F-49100 Angers, France;
| | - Catalina Sanz
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
- Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain
| | - María Isidoro-García
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain; (M.C.P.-M.); (E.M.-V.); (B.G.-B.); (D.H.H.-J.); (M.J.G.-S.); (S.M.L.-H.); (M.I.-G.)
- Institute for Biomedical Research of Salamanca, 37007 Salamanca, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
3
|
Russell LE, Claw KG, Aagaard KM, Glass SM, Dasgupta K, Nez FL, Haimbaugh A, Maldonato BJ, Yadav J. Insights into pharmacogenetics, drug-gene interactions, and drug-drug-gene interactions. Drug Metab Rev 2024:1-19. [PMID: 39154360 DOI: 10.1080/03602532.2024.2385928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024]
Abstract
This review explores genetic contributors to drug interactions, known as drug-gene and drug-drug-gene interactions (DGI and DDGI, respectively). This article is part of a mini-review issue led by the International Society for the Study of Xenobiotics (ISSX) New Investigators Group. Pharmacogenetics (PGx) is the study of the impact of genetic variation on pharmacokinetics (PK), pharmacodynamics (PD), and adverse drug reactions. Genetic variation in pharmacogenes, including drug metabolizing enzymes and drug transporters, is common and can increase the risk of adverse drug events or contribute to reduced efficacy. In this review, we summarize clinically actionable genetic variants, and touch on methodologies such as genotyping patient DNA to identify genetic variation in targeted genes, and deep mutational scanning as a high-throughput in vitro approach to study the impact of genetic variation on protein function and/or expression in vitro. We highlight the utility of physiologically based pharmacokinetic (PBPK) models to integrate genetic and chemical inhibitor and inducer data for more accurate human PK simulations. Additionally, we analyze the limitations of historical ethnic descriptors in pharmacogenomics research. Altogether, the work herein underscores the importance of identifying and understanding complex DGI and DDGIs with the intention to provide better treatment outcomes for patients. We also highlight current barriers to wide-scale implementation of PGx-guided dosing as standard or care in clinical settings.
Collapse
Affiliation(s)
- Laura E Russell
- Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| | - Katrina G Claw
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kaja M Aagaard
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah M Glass
- Preclinical Sciences and Translational Safety, Janssen Research &Development, San Diego, CA, USA
| | - Kuheli Dasgupta
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - F Leah Nez
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alex Haimbaugh
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc, Redwood City, CA, USA
| | - Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Boston, MA, USA
| |
Collapse
|
4
|
Pereira NL, Cresci S, Angiolillo DJ, Batchelor W, Capers Q, Cavallari LH, Leifer D, Luzum JA, Roden DM, Stellos K, Turrise SL, Tuteja S. CYP2C19 Genetic Testing for Oral P2Y12 Inhibitor Therapy: A Scientific Statement From the American Heart Association. Circulation 2024; 150:e129-e150. [PMID: 38899464 PMCID: PMC11300169 DOI: 10.1161/cir.0000000000001257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
There is significant variability in the efficacy and safety of oral P2Y12 inhibitors, which are used to prevent ischemic outcomes in common diseases such as coronary and peripheral arterial disease and stroke. Clopidogrel, a prodrug, is the most used oral P2Y12 inhibitor and is activated primarily after being metabolized by a highly polymorphic hepatic cytochrome CYP2C219 enzyme. Loss-of-function genetic variants in CYP2C219 are common, can result in decreased active metabolite levels and increased on-treatment platelet aggregation, and are associated with increased ischemic events on clopidogrel therapy. Such patients can be identified by CYP2C19 genetic testing and can be treated with alternative therapy. Conversely, universal use of potent oral P2Y12 inhibitors such as ticagrelor or prasugrel, which are not dependent on CYP2C19 for activation, has been recommended but can result in increased bleeding. Recent clinical trials and meta-analyses have demonstrated that a precision medicine approach in which loss-of-function carriers are prescribed ticagrelor or prasugrel and noncarriers are prescribed clopidogrel results in reducing ischemic events without increasing bleeding risk. The evidence to date supports CYP2C19 genetic testing before oral P2Y12 inhibitors are prescribed in patients with acute coronary syndromes or percutaneous coronary intervention. Clinical implementation of such genetic testing will depend on among multiple factors: rapid availability of results or adoption of the concept of performing preemptive genetic testing, provision of easy-to-understand results with therapeutic recommendations, and seamless integration in the electronic health record.
Collapse
|
5
|
Maes S, Deploey N, Peelman F, Eyckerman S. Deep mutational scanning of proteins in mammalian cells. CELL REPORTS METHODS 2023; 3:100641. [PMID: 37963462 PMCID: PMC10694495 DOI: 10.1016/j.crmeth.2023.100641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/06/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
Protein mutagenesis is essential for unveiling the molecular mechanisms underlying protein function in health, disease, and evolution. In the past decade, deep mutational scanning methods have evolved to support the functional analysis of nearly all possible single-amino acid changes in a protein of interest. While historically these methods were developed in lower organisms such as E. coli and yeast, recent technological advancements have resulted in the increased use of mammalian cells, particularly for studying proteins involved in human disease. These advancements will aid significantly in the classification and interpretation of variants of unknown significance, which are being discovered at large scale due to the current surge in the use of whole-genome sequencing in clinical contexts. Here, we explore the experimental aspects of deep mutational scanning studies in mammalian cells and report the different methods used in each step of the workflow, ultimately providing a useful guide toward the design of such studies.
Collapse
Affiliation(s)
- Stefanie Maes
- VIB Center for Medical Biotechnology (CMB), Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Nick Deploey
- VIB Center for Medical Biotechnology (CMB), Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Frank Peelman
- VIB Center for Medical Biotechnology (CMB), Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Sven Eyckerman
- VIB Center for Medical Biotechnology (CMB), Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium.
| |
Collapse
|
6
|
Tremmel R, Pirmann S, Zhou Y, Lauschke VM. Translating pharmacogenomic sequencing data into drug response predictions-How to interpret variants of unknown significance. Br J Clin Pharmacol 2023. [PMID: 37759374 DOI: 10.1111/bcp.15915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
The rapid development of sequencing technologies during the past 20 years has provided a variety of methods and tools to interrogate human genomic variations at the population level. Pharmacogenes are well known to be highly polymorphic and a plethora of pharmacogenomic variants has been identified in population sequencing data. However, so far only a small number of these variants have been functionally characterized regarding their impact on drug efficacy and toxicity and the significance of the vast majority remains unknown. It is therefore of high importance to develop tools and frameworks to accurately infer the effects of pharmacogenomic variants and, eventually, aggregate the effect of individual variations into personalized drug response predictions. To address this challenge, we here first describe the technological advances, including sequencing methods and accompanying bioinformatic processing pipelines that have enabled reliable variant identification. Subsequently, we highlight advances in computational algorithms for pharmacogenomic variant interpretation and discuss the added value of emerging strategies, such as machine learning and the integrative use of omics techniques that have the potential to further contribute to the refinement of personalized pharmacological response predictions. Lastly, we provide an overview of experimental and clinical approaches to validate in silico predictions. We conclude that the iterative feedback between computational predictions and experimental validations is likely to rapidly improve the accuracy of pharmacogenomic prediction models, which might soon allow for an incorporation of the entire pharmacogenetic profile into personalized response predictions.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Sebastian Pirmann
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Information and Data Science School for Health, Karlsruhe/Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Khoza N, Twesigomwe D, Othman H. Characterizing the combined effects of cytochrome P450 missense variation within star allele definitions. Pharmacogenomics 2023; 24:561-578. [PMID: 37503750 DOI: 10.2217/pgs-2023-0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Background: Cytochrome P450 (CYP) genetic variation largely impacts drug response. However, many CYP star alleles (haplotypes) lack functional annotation, impeding our understanding of drug metabolism mechanisms. We aimed to investigate the impact of missense variant combinations on CYP protein structures. Methods: Normal mode analysis was conducted on 261 missense variants within 91 CYP haplotypes. CYP2D6*2 and CYP2D6*17 were prioritized for molecular dynamics simulation. Results: Normal mode analysis and molecular dynamics highlight the effects of known CYP missense variants on protein stability and conformational dynamics. Missense variants within haplotypes may have intermodulating effects on protein structure and function. Conclusion: This study highlights the utility of multiscale modeling in interpreting CYP missense variants and particularly their combinations within various star alleles.
Collapse
Affiliation(s)
- Nhlamulo Khoza
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, 9 Jubilee Road, Parktown, Johannesburg, 2193, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2001, South Africa
| | - David Twesigomwe
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, 9 Jubilee Road, Parktown, Johannesburg, 2193, South Africa
| | - Houcemeddine Othman
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, 9 Jubilee Road, Parktown, Johannesburg, 2193, South Africa
- Laboratory of Human Cytogenetics, Molecular Genetics and Reproductive Biology, Farhat Hached University Hospital, Sousse, 4000, Tunisia
| |
Collapse
|
8
|
Abstract
Antiplatelet therapy is used in the treatment of patients with acute coronary syndromes, stroke, and those undergoing percutaneous coronary intervention. Clopidogrel is the most widely used antiplatelet P2Y12 inhibitor in clinical practice. Genetic variation in CYP2C19 may influence its enzymatic activity, resulting in individuals who are carriers of loss-of-function CYP2C19 alleles and thus have reduced active clopidogrel metabolites, high on-treatment platelet reactivity, and increased ischemic risk. Prospective studies have examined the utility of CYP2C19 genetic testing to guide antiplatelet therapy, and more recently published meta-analyses suggest that pharmacogenetics represents a key treatment strategy to individualize antiplatelet therapy. Rapid genetic tests, including bedside genotyping platforms that are validated and have high reproducibility, are available to guide selection of P2Y12 inhibitors in clinical practice. The aim of this review is to provide an overview of the background and rationale for the role of a guided antiplatelet approach to enhance patient care.
Collapse
Affiliation(s)
- Matteo Castrichini
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA;
| | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan, USA
| | - Naveen Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA;
| |
Collapse
|
9
|
Seo ME, Min BJ, Heo N, Lee KH, Kim JH. Comprehensive in vitro and in silico assessments of metabolic capabilities of 24 genomic variants of CYP2C19 using two different substrates. Front Pharmacol 2023; 14:1055991. [PMID: 36713839 PMCID: PMC9877350 DOI: 10.3389/fphar.2023.1055991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Introduction: Most hepatically cleared drugs are metabolized by cytochromes P450 (CYPs), and Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines provide curated clinical references for CYPs to apply individual genome data for optimized drug therapy. However, incorporating novel pharmacogenetic variants into guidelines takes considerable time. Methods: We comprehensively assessed the drug metabolizing capabilities of CYP2C19 variants discovered through population sequencing of two substrates, S-mephenytoin and omeprazole. Results: Based on established functional assays, 75% (18/24) of the variants not yet described in Pharmacogene Variation (PharmVar) had significantly altered drug metabolizing capabilities. Of them, seven variants with inappreciable protein expression were evaluated as protein damaging by all three in silico prediction algorithms, Sorting intolerant from tolerant (SIFT), Polymorphism Phenotyping v2 (PolyPhen-2), and Combined annotation dependent depletion (CADD). The five variants with decreased metabolic capability (<50%) of wild type for either substrates were evaluated as protein damaging by all three in silico prediction algorithms, except CADD exact score of NM_000769.4:c.593T>C that was 19.68 (<20.0). In the crystal structure of the five polymorphic proteins, each altered residue of all those proteins was observed to affect the key structures of drug binding specificity. We also identified polymorphic proteins indicating different tendencies of metabolic capability between the two substrates (5/24). Discussion: Therefore, we propose a methodology that combines in silico prediction algorithms and functional assays on polymorphic CYPs with multiple substrates to evaluate the changes in the metabolism of all possible genomic variants in CYP genes. The approach would reinforce existing guidelines and provide information for prescribing appropriate medicines for individual patients.
Collapse
Affiliation(s)
- Myung-Eui Seo
- Seoul National University Biomedical Informatics (SNUBI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Byung-Joo Min
- National Forensic Service Seoul Institute, Seoul, South Korea
| | - Nayoon Heo
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kye Hwa Lee
- Department of Information Medicine, Asan Medical Center and University of Ulsan College of Medicine, Seoul, South Korea,*Correspondence: Kye Hwa Lee, ; Ju Han Kim,
| | - Ju Han Kim
- Seoul National University Biomedical Informatics (SNUBI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea,Seoul National University Biomedical Informatics (SNUBI), Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, South Korea,*Correspondence: Kye Hwa Lee, ; Ju Han Kim,
| |
Collapse
|
10
|
Zhou Y, Lauschke VM. Challenges Related to the Use of Next-Generation Sequencing for the Optimization of Drug Therapy. Handb Exp Pharmacol 2023; 280:237-260. [PMID: 35792943 DOI: 10.1007/164_2022_596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Over the last decade, next-generation sequencing (NGS) methods have become increasingly used in various areas of human genomics. In routine clinical care, their use is already implemented in oncology to profile the mutational landscape of a tumor, as well as in rare disease diagnostics. However, its utilization in pharmacogenomics is largely lacking behind. Recent population-scale genome data has revealed that human pharmacogenes carry a plethora of rare genetic variations that are not interrogated by conventional array-based profiling methods and it is estimated that these variants could explain around 30% of the genetically encoded functional pharmacogenetic variability.To interpret the impact of such variants on drug response a multitude of computational tools have been developed, but, while there have been major advancements, it remains to be shown whether their accuracy is sufficient to improve personalized pharmacogenetic recommendations in robust trials. In addition, conventional short-read sequencing methods face difficulties in the interrogation of complex pharmacogenes and high NGS test costs require stringent evaluations of cost-effectiveness to decide about reimbursement by national healthcare programs. Here, we illustrate current challenges and discuss future directions toward the clinical implementation of NGS to inform genotype-guided decision-making.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.
- University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
11
|
Fu Y, Bedő J, Papenfuss AT, Rubin AF. Integrating deep mutational scanning and low-throughput mutagenesis data to predict the impact of amino acid variants. Gigascience 2022; 12:giad073. [PMID: 37721410 PMCID: PMC10506130 DOI: 10.1093/gigascience/giad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/02/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND Evaluating the impact of amino acid variants has been a critical challenge for studying protein function and interpreting genomic data. High-throughput experimental methods like deep mutational scanning (DMS) can measure the effect of large numbers of variants in a target protein, but because DMS studies have not been performed on all proteins, researchers also model DMS data computationally to estimate variant impacts by predictors. RESULTS In this study, we extended a linear regression-based predictor to explore whether incorporating data from alanine scanning (AS), a widely used low-throughput mutagenesis method, would improve prediction results. To evaluate our model, we collected 146 AS datasets, mapping to 54 DMS datasets across 22 distinct proteins. CONCLUSIONS We show that improved model performance depends on the compatibility of the DMS and AS assays, and the scale of improvement is closely related to the correlation between DMS and AS results.
Collapse
Affiliation(s)
- Yunfan Fu
- The Walter and Eliza Hall Institute of Medical Research, Bioinformatics Division, 1G Royal Pde, Parkville, Victoria 3052, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, Victoria 3010, Australia
| | - Justin Bedő
- The Walter and Eliza Hall Institute of Medical Research, Bioinformatics Division, 1G Royal Pde, Parkville, Victoria 3052, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, Victoria 3010, Australia
| | - Anthony T Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Bioinformatics Division, 1G Royal Pde, Parkville, Victoria 3052, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, Victoria 3010, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Alan F Rubin
- The Walter and Eliza Hall Institute of Medical Research, Bioinformatics Division, 1G Royal Pde, Parkville, Victoria 3052, Australia
- The University of Melbourne, Department of Medical Biology, Parkville, Victoria 3010, Australia
| |
Collapse
|
12
|
Zhao FL, Zhang Q, Wang SH, Hong Y, Zhou S, Zhou Q, Geng PW, Luo QF, Yang JF, Chen H, Cai JP, Dai DP. Identification and drug metabolic characterization of four new CYP2C9 variants CYP2C9*72- *75 in the Chinese Han population. Front Pharmacol 2022; 13:1007268. [PMID: 36582532 PMCID: PMC9792615 DOI: 10.3389/fphar.2022.1007268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Cytochrome 2C9 (CYP2C9), one of the most important drug metabolic enzymes in the human hepatic P450 superfamily, is required for the metabolism of 15% of clinical drugs. Similar to other CYP2C family members, CYP2C9 gene has a high genetic polymorphism which can cause significant racial and inter-individual differences in drug metabolic activity. To better understand the genetic distribution pattern of CYP2C9 in the Chinese Han population, 931 individuals were recruited and used for the genotyping in this study. As a result, seven synonymous and 14 non-synonymous variations were identified, of which 4 missense variants were designated as new alleles CYP2C9*72, *73, *74 and *75, resulting in the amino acid substitutions of A149V, R150C, Q214H and N418T, respectively. When expressed in insect cell microsomes, all four variants exhibited comparable protein expression levels to that of the wild-type CYP2C9 enzyme. However, drug metabolic activity analysis revealed that these variants exhibited significantly decreased catalytic activities toward three CYP2C9 specific probe drugs, as compared with that of the wild-type enzyme. These data indicate that the amino acid substitution in newly designated variants can cause reduced function of the enzyme and its clinical significance still needs further investigation in the future.
Collapse
Affiliation(s)
- Fang-Ling Zhao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China,Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Qing Zhang
- Department of Cardiovascular, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuang-Hu Wang
- Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Yun Hong
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shan Zhou
- Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Quan Zhou
- Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Pei-Wu Geng
- Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Qing-Feng Luo
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie-Fu Yang
- Department of Cardiovascular, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Chen
- Department of Cardiovascular, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China,*Correspondence: Da-Peng Dai, ; Jian-Ping Cai, ; Hao Chen,
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China,*Correspondence: Da-Peng Dai, ; Jian-Ping Cai, ; Hao Chen,
| | - Da-Peng Dai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China,Peking University Fifth School of Clinical Medicine, Beijing, China,*Correspondence: Da-Peng Dai, ; Jian-Ping Cai, ; Hao Chen,
| |
Collapse
|
13
|
Tabet D, Parikh V, Mali P, Roth FP, Claussnitzer M. Scalable Functional Assays for the Interpretation of Human Genetic Variation. Annu Rev Genet 2022; 56:441-465. [PMID: 36055970 DOI: 10.1146/annurev-genet-072920-032107] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Scalable sequence-function studies have enabled the systematic analysis and cataloging of hundreds of thousands of coding and noncoding genetic variants in the human genome. This has improved clinical variant interpretation and provided insights into the molecular, biophysical, and cellular effects of genetic variants at an astonishing scale and resolution across the spectrum of allele frequencies. In this review, we explore current applications and prospects for the field and outline the principles underlying scalable functional assay design, with a focus on the study of single-nucleotide coding and noncoding variants.
Collapse
Affiliation(s)
- Daniel Tabet
- Donnelly Centre, Department of Molecular Genetics, and Department of Computer Science, University of Toronto, Toronto, Ontario, Canada;
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Victoria Parikh
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, California, USA
| | - Frederick P Roth
- Donnelly Centre, Department of Molecular Genetics, and Department of Computer Science, University of Toronto, Toronto, Ontario, Canada;
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA;
| |
Collapse
|
14
|
An L, Wang Y, Wu G, Wang Z, Shi Z, Liu C, Wang C, Yi M, Niu C, Duan S, Li X, Tang W, Wu K, Chen S, Xu H. Defining the sensitivity landscape of EGFR variants to tyrosine kinase inhibitors. Transl Res 2022; 255:14-25. [PMID: 36347492 DOI: 10.1016/j.trsl.2022.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/06/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022]
Abstract
Tyrosine kinase inhibitor (TKI) is a standard treatment for patients with NSCLC harboring constitutively active epidermal growth factor receptor (EGFR) mutations. However, most rare EGFR mutations lack treatment regimens except for the well-studied ones. We constructed two EGFR variant libraries containing substitutions, deletions, or insertions using the saturation mutagenesis method. All the variants were located in the EGFR mutation hotspot (exons 18-21). The sensitivity of these variants to afatinib, erlotinib, gefitinib, icotinib, and osimertinib was systematically studied by determining their enrichment in massively parallel cytotoxicity assays using an endogenous EGFR-depleted cell line. A total of 3914 and 70,475 variants were detected in the constructed EGFR Substitution-Deletion (Sub-Del) and exon 20 Insertion (Ins) libraries. Of the 3914 Sub-Del variants, 221 proliferated fast in the control assay and were sensitive to EGFR-TKIs. For the 70,475 Ins variants, insertions at amino acid positions 770-774 were highly enriched in all 5 TKI cytotoxicity assays. Moreover, the top 5% of the enriched insertion variants included a glycine or serine insertion at high frequency. We present a comprehensive reference for the sensitivity of EGFR variants to five commonly used TKIs. The approach used here should be applicable to other genes and targeted drugs. BACKGROUND: Tyrosine kinase inhibitors (TKIs) therapy is a standard treatment for patients with advanced non-small-cell lung carcinoma (NSCLC) when activating epidermal growth factor receptor (EGFR) mutations are detected. However, except for the well-studied EGFR mutations, most EGFR mutations lack treatment regimens. TRANSLATIONAL SIGNIFICANCE: The results demonstrated that patients with rare EGFR mutations were most likely to benefit from osimertinib therapy compared to afatinib, erlotinib, gefitinib, or icotinib therapy. This study provides a case of deep mutational scanning that simultaneously assayed substitution, deletion, and insertion variants. This approach is applicable for other oncogenes and targeted drugs.
Collapse
Affiliation(s)
- Lei An
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng 475000, China
| | | | - Guangyao Wu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng 475000, China
| | - Zhenxing Wang
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng 475000, China
| | - Zeyuan Shi
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng 475000, China
| | - Chang Liu
- School of Pharmacy, Henan University, Kaifeng 475000, China
| | - Chunli Wang
- School of Pharmacy, Henan University, Kaifeng 475000, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenguang Niu
- Key Laboratory of Clinical Resources Translation, The First Affiliated Hospital of Henan University, Kaifeng 475000, China
| | - Shaofeng Duan
- School of Pharmacy, Henan University, Kaifeng 475000, China
| | - Xiaodong Li
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng 475000, China
| | - Wenxue Tang
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China; The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuqing Chen
- Shenzhen Typhoon HealthCare, Shenzhen 518000, China.
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China; The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
15
|
Zhou Y, Tremmel R, Schaeffeler E, Schwab M, Lauschke VM. Challenges and opportunities associated with rare-variant pharmacogenomics. Trends Pharmacol Sci 2022; 43:852-865. [PMID: 36008164 DOI: 10.1016/j.tips.2022.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/15/2022] [Accepted: 07/29/2022] [Indexed: 12/26/2022]
Abstract
Recent advances in next-generation sequencing (NGS) have resulted in the identification of tens of thousands of rare pharmacogenetic variations with unknown functional effects. However, although such pharmacogenetic variations have been estimated to account for a considerable amount of the heritable variability in drug response and toxicity, accurate interpretation at the level of the individual patient remains challenging. We discuss emerging strategies and concepts to close this translational gap. We illustrate how massively parallel experimental assays, artificial intelligence (AI), and machine learning can synergize with population-scale biobank projects to facilitate the interpretation of NGS data to individualize clinical decision-making and personalized medicine.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Roman Tremmel
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tübingen, Tübingen, Germany
| | - Elke Schaeffeler
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Cluster of Excellence iFIT (EXC2180) Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany; Department of Clinical Pharmacology, and Department of Biochemistry and Pharmacy, University of Tübingen, Tübingen, Germany
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden; Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tübingen, Tübingen, Germany.
| |
Collapse
|
16
|
Zhang L, Sarangi V, Liu D, Ho MF, Grassi AR, Wei L, Moon I, Vierkant RA, Larson NB, Lazaridis KN, Athreya AP, Wang L, Weinshilboum R. ACE2 and TMPRSS2 SARS-CoV-2 infectivity genes: deep mutational scanning and characterization of missense variants. Hum Mol Genet 2022; 31:4183-4192. [PMID: 35861636 PMCID: PMC9759330 DOI: 10.1093/hmg/ddac157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/18/2022] [Accepted: 07/05/2022] [Indexed: 01/21/2023] Open
Abstract
The human angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) proteins play key roles in the cellular internalization of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus responsible for the coronavirus disease of 2019 (COVID-19) pandemic. We set out to functionally characterize the ACE2 and TMPRSS2 protein abundance for variant alleles encoding these proteins that contained non-synonymous single-nucleotide polymorphisms (nsSNPs) in their open reading frames (ORFs). Specifically, a high-throughput assay, deep mutational scanning (DMS), was employed to test the functional implications of nsSNPs, which are variants of uncertain significance in these two genes. Specifically, we used a 'landing pad' system designed to quantify the protein expression for 433 nsSNPs that have been observed in the ACE2 and TMPRSS2 ORFs and found that 8 of 127 ACE2, 19 of 157 TMPRSS2 isoform 1 and 13 of 149 TMPRSS2 isoform 2 variant proteins displayed less than ~25% of the wild-type protein expression, whereas 4 ACE2 variants displayed 25% or greater increases in protein expression. As a result, we concluded that nsSNPs in genes encoding ACE2 and TMPRSS2 might potentially influence SARS-CoV-2 infectivity. These results can now be applied to DNA sequence data for patients infected with SARS-CoV-2 to determine the possible impact of patient-based DNA sequence variation on the clinical course of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lingxin Zhang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Vivekananda Sarangi
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Angela R Grassi
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Lixuan Wei
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Irene Moon
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert A Vierkant
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicholas B Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA,Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Arjun P Athreya
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA,Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA,Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard Weinshilboum
- To whom correspondence should be addressed at: Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic 200 First Street SW, Rochester, MN 55905, USA. Tel: +1 5072842246;
| |
Collapse
|
17
|
Silva-Alarcon S, Valencia C, Newball L, Saldarriaga W, Castillo A. Molecular Variants in Genes related to the Response to Ocular Hypotensive Drugs in an Afro-Colombian Population. Open Ophthalmol J 2022. [DOI: 10.2174/18743641-v16-e2205250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aims:
This study aimed to conduct an exploratory analysis of the pharmacogenomic variants involved in ocular hypotensive drugs to understand the individual differential response in an Afro-descendant population.
Background:
Glaucoma is the leading cause of irreversible blindness worldwide. The pharmacologic treatment available consists of lowering intraocular pressure by administering topical drugs. In Asian and Caucasian people, pharmacogenomic variants associated with the efficacy of these treatments have been identified. However, in Afro-descendant populations, there is a profound gap in this knowledge.
Objective:
This study identified the pharmacogenomic variants related to ocular hypotensive efficacy treatment in Afro-descendant individuals from the Archipelago of San Andres and Providence, Colombia.
Methods:
An analysis of whole-exome sequencings (WES), functional annotation, and clinical significance was performed for pharmacogenomic variants reported in PharmGKB databases; in turn, an in silico available prediction analysis was carried out for the novel variants.
Results:
We identified six out of 18 non-synonymous variants with a clinical annotation in PharmGKB. Five were classified as level three evidence for the hypotensive drugs; rs1801252 and rs1801253 in the ADRB1 gene and rs1042714 in the ADRB2 gene. These pharmacogenomic variants have been involved in a lack of efficacy of topical beta-blockers and higher systolic and diastolic pressure under treatment with ophthalmic timolol drug. The rs1045642 in the ABCB1 gene was associated with greater efficacy of treatments with latanoprost drug. Also, we found the haplotypes *17 for CYP2D6 and *10 for CYP2C19; both related to reducing the enzyme activity to timolol drug metabolization. In addition, we observed 50 novel potentially actionable variants; 36 synonymous, two insertion variants that caused frameshift mutations, and 12 non-synonymous, where five were predicted to be pathogenic based on several pathogenicity predictions.
Conclusion:
Our results suggested that the pharmacogenomic variants were found to decrease the ocular hypotensive efficacy treatment in a Colombian Afro-descendant population and revealed a significant proportion of novel variants with a potential to influence drug response.
Collapse
|
18
|
Wang L, Scherer SE, Bielinski SJ, Muzny DM, Jones LA, Black JL, Moyer AM, Giri J, Sharp RR, Matey ET, Wright JA, Oyen LJ, Nicholson WT, Wiepert M, Sullard T, Curry TB, Vitek CRR, McAllister TM, Sauver JL, Caraballo PJ, Lazaridis KN, Venner E, Qin X, Hu J, Kovar CL, Korchina V, Walker K, Doddapaneni H, Wu TJ, Raj R, Denson S, Liu W, Chandanavelli G, Zhang L, Wang Q, Kalra D, Karow MB, Harris KJ, Sicotte H, Peterson SE, Barthel AE, Moore BE, Skierka JM, Kluge ML, Kotzer KE, Kloke K, Vander Pol JM, Marker H, Sutton JA, Kekic A, Ebenhoh A, Bierle DM, Schuh MJ, Grilli C, Erickson S, Umbreit A, Ward L, Crosby S, Nelson EA, Levey S, Elliott M, Peters SG, Pereira N, Frye M, Shamoun F, Goetz MP, Kullo IJ, Wermers R, Anderson JA, Formea CM, El Melik RM, Zeuli JD, Herges JR, Krieger CA, Hoel RW, Taraba JL, Thomas SR, Absah I, Bernard ME, Fink SR, Gossard A, Grubbs PL, Jacobson TM, Takahashi P, Zehe SC, Buckles S, Bumgardner M, Gallagher C, Fee-Schroeder K, Nicholas NR, Powers ML, Ragab AK, Richardson DM, Stai A, Wilson J, Pacyna JE, Olson JE, Sutton EJ, Beck AT, Horrow C, Kalari KR, Larson NB, Liu H, Wang L, Lopes GS, Borah BJ, Freimuth RR, Zhu Y, Jacobson DJ, Hathcock MA, Armasu SM, McGree ME, Jiang R, Koep TH, Ross JL, Hilden M, Bosse K, Ramey B, Searcy I, Boerwinkle E, Gibbs RA, Weinshilboum RM. Implementation of preemptive DNA sequence-based pharmacogenomics testing across a large academic medical center: The Mayo-Baylor RIGHT 10K Study. Genet Med 2022; 24:1062-1072. [PMID: 35331649 PMCID: PMC9272414 DOI: 10.1016/j.gim.2022.01.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The Mayo-Baylor RIGHT 10K Study enabled preemptive, sequence-based pharmacogenomics (PGx)-driven drug prescribing practices in routine clinical care within a large cohort. We also generated the tools and resources necessary for clinical PGx implementation and identified challenges that need to be overcome. Furthermore, we measured the frequency of both common genetic variation for which clinical guidelines already exist and rare variation that could be detected by DNA sequencing, rather than genotyping. METHODS Targeted oligonucleotide-capture sequencing of 77 pharmacogenes was performed using DNA from 10,077 consented Mayo Clinic Biobank volunteers. The resulting predicted drug response-related phenotypes for 13 genes, including CYP2D6 and HLA, affecting 21 drug-gene pairs, were deposited preemptively in the Mayo electronic health record. RESULTS For the 13 pharmacogenes of interest, the genomes of 79% of participants carried clinically actionable variants in 3 or more genes, and DNA sequencing identified an average of 3.3 additional conservatively predicted deleterious variants that would not have been evident using genotyping. CONCLUSION Implementation of preemptive rather than reactive and sequence-based rather than genotype-based PGx prescribing revealed nearly universal patient applicability and required integrated institution-wide resources to fully realize individualized drug therapy and to show more efficient use of health care resources.
Collapse
Affiliation(s)
- Liewei Wang
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN,Division of Clinical Pharmacology, Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Steven E. Scherer
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Suzette J. Bielinski
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Donna M. Muzny
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Leila A. Jones
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - John Logan Black
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ann M. Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Jyothsna Giri
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | | | - Wayne T. Nicholson
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Mathieu Wiepert
- Department of Information Technology, Mayo Clinic, Rochester, MN
| | - Terri Sullard
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Timothy B. Curry
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Jennifer L. Sauver
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN,Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Pedro J. Caraballo
- Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Konstantinos N. Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN,Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Eric Venner
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Xiang Qin
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Jianhong Hu
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Christie L. Kovar
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Viktoriya Korchina
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Kimberly Walker
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | | | - Tsung-Jung Wu
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Ritika Raj
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Shawn Denson
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Wen Liu
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Gauthami Chandanavelli
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Lan Zhang
- Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX
| | - Qiaoyan Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Divya Kalra
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Mary Beth Karow
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Hugues Sicotte
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Sandra E. Peterson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Amy E. Barthel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Brenda E. Moore
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Michelle L. Kluge
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Katrina E. Kotzer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Karen Kloke
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Heather Marker
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Joseph A. Sutton
- Department of Information Technology, Mayo Clinic, Rochester, MN
| | | | | | - Dennis M. Bierle
- Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | - Audrey Umbreit
- Department of Pharmacy, Mayo Clinic Health System, Mankato, MN
| | - Leah Ward
- Department of Pharmacy, Mayo Clinic, Jacksonville, FL
| | - Sheena Crosby
- Department of Pharmacy, Mayo Clinic, Jacksonville, FL
| | | | - Sharon Levey
- Department of Clinical Genomics, Mayo Clinic, Scottsdale, AZ
| | - Michelle Elliott
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Steve G. Peters
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Naveen Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Mark Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN
| | - Fadi Shamoun
- Department of Cardiovascular Medicine Mayo Clinic, Phoenix, AZ
| | - Matthew P. Goetz
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN
| | | | - Robert Wermers
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | - Scott R. Thomas
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Imad Absah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | - Stephanie R. Fink
- Division of Community Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Andrea Gossard
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Paul Takahashi
- Division of Community Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | - Susan Buckles
- Department of Public Affairs, Mayo Clinic, Rochester, MN
| | | | | | | | | | - Melody L. Powers
- Biospecimens Accessioning and Processing Laboratory, Mayo Clinic, Rochester, MN
| | - Ahmed K. Ragab
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Anthony Stai
- Department of Information Technology, Mayo Clinic, Rochester, MN
| | - Jaymi Wilson
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Joel E. Pacyna
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN
| | - Janet E. Olson
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN,Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN
| | - Erica J. Sutton
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN
| | - Annika T. Beck
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN
| | - Caroline Horrow
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN
| | - Krishna R. Kalari
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Nicholas B. Larson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Hongfang Liu
- Division of Digital Health Sciences, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Liwei Wang
- Division of Digital Health Sciences, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Guilherme S. Lopes
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN,Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Bijan J. Borah
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN,Division of Health Care Policy and Research, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Robert R. Freimuth
- Division of Digital Health Sciences, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Ye Zhu
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Debra J. Jacobson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Matthew A. Hathcock
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Sebastian M. Armasu
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Michaela E. McGree
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Ruoxiang Jiang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX,Human Genome Sequencing Center Clinical Laboratory, Baylor College of Medicine, Houston, TX,School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX,Corresponding Authors (), ()
| | - Richard M. Weinshilboum
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN,Division of Clinical Pharmacology, Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN,Corresponding Authors (), ()
| |
Collapse
|
19
|
Brown KE, Staples JW, Woodahl EL. Keeping pace with CYP2D6 haplotype discovery: innovative methods to assign function. Pharmacogenomics 2022; 23:255-262. [PMID: 35083931 PMCID: PMC8890136 DOI: 10.2217/pgs-2021-0149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The discovery of haplotypes with unknown or uncertain function in the CYP2D6 pharmacogene is outpacing the capabilities of traditional in vitro and in vivo approaches to characterize their function. This challenge will undoubtedly grow as pharmacogenomic research becomes more inclusive of globally diverse populations. As accurate phenotypic assignment is paramount to the utility of pharmacogenomics, high-throughput technologies are needed for this complex pharmacogene. We describe the evolving landscape of innovative approaches to assign function to CYP2D6 haplotypes and possibilities for adopting these technologies into cohesive processes. Promising approaches include ADME-optimized prediction frameworks, machine learning algorithms, deep mutational scanning and phenoconversion predictions. Implementing these approaches will lead to improved personalization of treatment for patients.
Collapse
Affiliation(s)
- Karen E Brown
- Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, USA,Skaggs Institute for Health Innovation, University of Montana, Missoula, MT 59812, USA
| | - Jack W Staples
- Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, USA,Skaggs Institute for Health Innovation, University of Montana, Missoula, MT 59812, USA
| | - Erica L Woodahl
- Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, USA,Skaggs Institute for Health Innovation, University of Montana, Missoula, MT 59812, USA,Author for correspondence: Tel.: +1 406 243 4129;
| |
Collapse
|
20
|
Lopes JL, Harris K, Karow MB, Peterson SE, Kluge ML, Kotzer KE, Lopes GS, Larson NB, Bielinski SJ, Scherer SE, Wang L, Weinshilboum RM, Black JL, Moyer AM. Targeted Genotyping in Clinical Pharmacogenomics: What Is Missing? J Mol Diagn 2022; 24:253-261. [PMID: 35041929 PMCID: PMC8961466 DOI: 10.1016/j.jmoldx.2021.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Clinical pharmacogenomic testing typically uses targeted genotyping, which only detects variants included in the test design and may vary among laboratories. To evaluate the potential patient impact of genotyping compared with sequencing, which can detect common and rare variants, an in silico targeted genotyping panel was developed based on the variants most commonly included in clinical tests and applied to a cohort of 10,030 participants who underwent sequencing for CYP1A2, CYP2C19, CYP2C9, CYP2D6, CYP3A4, CYP3A5, DPYD, SLCO1B1, TPMT, UGT1A1, and VKORC1. The results of in silico targeted genotyping were compared with the clinically reported sequencing results. Of the 10,030 participants, 2780 (28%) had at least one potentially clinically relevant variant/allele identified by sequencing that would not have been detected in a standard targeted genotyping panel. The genes with the largest number of participants with variants only detected by sequencing were SLCO1B1, DPYD, and CYP2D6, which affected 13%, 6.3%, and 3.5% of participants, respectively. DPYD (112 variants) and CYP2D6 (103 variants) had the largest number of unique variants detected only by sequencing. Although targeted genotyping detects most clinically significant pharmacogenomic variants, sequencing-based approaches are necessary to detect rare variants that collectively affect many patients. However, efforts to establish pharmacogenomic variant classification systems and nomenclature to accommodate rare variants will be required to adopt sequencing-based pharmacogenomics.
Collapse
Affiliation(s)
- Jaime L. Lopes
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kimberley Harris
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Mary Beth Karow
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Sandra E. Peterson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Michelle L. Kluge
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Katrina E. Kotzer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Guilherme S. Lopes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Nicholas B. Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | | | - Steven E. Scherer
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - John L. Black
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Ann M. Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota,Address correspondence to Ann M. Moyer, M.D., Ph.D., Mayo Clinic, 200 First St SW, Rochester, MN 55905.
| |
Collapse
|
21
|
Powell NR, Zhao H, Ipe J, Liu Y, Skaar TC. Mapping the miRNA-mRNA Interactome in Human Hepatocytes and Identification of Functional mirSNPs in Pharmacogenes. Clin Pharmacol Ther 2021; 110:1106-1118. [PMID: 34314509 PMCID: PMC9007393 DOI: 10.1002/cpt.2379] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022]
Abstract
MiRNAs regulate the expression of hepatic genes involved in pharmacokinetics and pharmacodynamics. Genetic variants affecting miRNA binding (mirSNPs) have been associated with altered drug response, but previously used methods to identify miRNA binding sites and functional mirSNPs in pharmacogenes are indirect and limited by low throughput. We utilized the high-throughput chimeric-eCLIP assay to directly map thousands of miRNA-mRNA interactions and define the miRNA binding sites in primary hepatocytes. We then used the high-throughput PASSPORT-seq assay to functionally test 262 potential mirSNPs with coordinates overlapping the identified miRNA binding sites. Using chimeric-eCLIP, we identified a network of 448 miRNAs that collectively target 11,263 unique genes in primary hepatocytes pooled from 100 donors. Our data provide an extensive map of miRNA binding of each gene, including pharmacogenes, expressed in primary hepatocytes. For example, we identified the hsa-mir-27b-DPYD interaction at a previously validated binding site. A second example is our identification of 19 unique miRNAs that bind to CYP2B6 across 20 putative binding sites on the transcript. Using PASSPORT-seq, we then identified 24 mirSNPs that functionally impacted reporter mRNA levels. To our knowledge, this is the most comprehensive identification of miRNA binding sites in pharmacogenes. Combining chimeric-eCLIP with PASSPORT-seq successfully identified functional mirSNPs in pharmacogenes that may affect transcript levels through altered miRNA binding. These results provide additional insights into potential mechanisms contributing to interindividual variability in drug response.
Collapse
Affiliation(s)
- Nicholas R. Powell
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, Indiana, USA
| | - Harrison Zhao
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, Indianapolis, Indiana, USA
| | - Joseph Ipe
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, Indiana, USA
| | - Yunlong Liu
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, Indianapolis, Indiana, USA
| | - Todd C. Skaar
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, Indiana, USA
| |
Collapse
|
22
|
Geck RC, Boyle G, Amorosi CJ, Fowler DM, Dunham MJ. Measuring Pharmacogene Variant Function at Scale Using Multiplexed Assays. Annu Rev Pharmacol Toxicol 2021; 62:531-550. [PMID: 34516287 DOI: 10.1146/annurev-pharmtox-032221-085807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As costs of next-generation sequencing decrease, identification of genetic variants has far outpaced our ability to understand their functional consequences. This lack of understanding is a central challenge to a key promise of pharmacogenomics: using genetic information to guide drug selection and dosing. Recently developed multiplexed assays of variant effect enable experimental measurement of the function of thousands of variants simultaneously. Here, we describe multiplexed assays that have been performed on nearly 25,000 variants in eight key pharmacogenes (ADRB2, CYP2C9, CYP2C19, NUDT15, SLCO1B1, TMPT, VKORC1, and the LDLR promoter), discuss advances in experimental design, and explore key challenges that must be overcome to maximize the utility of multiplexed functional data. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Renee C Geck
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA; ,
| | - Gabriel Boyle
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA; ,
| | - Clara J Amorosi
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA; ,
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA; , .,Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA
| | - Maitreya J Dunham
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA; ,
| |
Collapse
|
23
|
Massively parallel characterization of CYP2C9 variant enzyme activity and abundance. Am J Hum Genet 2021; 108:1735-1751. [PMID: 34314704 DOI: 10.1016/j.ajhg.2021.07.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/28/2021] [Indexed: 12/19/2022] Open
Abstract
CYP2C9 encodes a cytochrome P450 enzyme responsible for metabolizing up to 15% of small molecule drugs, and CYP2C9 variants can alter the safety and efficacy of these therapeutics. In particular, the anti-coagulant warfarin is prescribed to over 15 million people annually and polymorphisms in CYP2C9 can affect individual drug response and lead to an increased risk of hemorrhage. We developed click-seq, a pooled yeast-based activity assay, to test thousands of variants. Using click-seq, we measured the activity of 6,142 missense variants in yeast. We also measured the steady-state cellular abundance of 6,370 missense variants in a human cell line by using variant abundance by massively parallel sequencing (VAMP-seq). These data revealed that almost two-thirds of CYP2C9 variants showed decreased activity and that protein abundance accounted for half of the variation in CYP2C9 function. We also measured activity scores for 319 previously unannotated human variants, many of which may have clinical relevance.
Collapse
|
24
|
Naushad SM, Kutala VK, Hussain T, Alrokayan SA. Pharmacogenetic determinants of warfarin in the Indian population. Pharmacol Rep 2021; 73:1396-1404. [PMID: 34106453 DOI: 10.1007/s43440-021-00297-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Several studies optimized the warfarin dose based on CYP2C9*2, CYP2C9*3, VKORC1 -1639 G > A, CYP4F2 V433M. But, the information on the rare variants is lacking. In this study, we have explored the prevalence of common and rare pharmacogenetic determinants of warfarin and determined their damaging nature. METHODS We have analyzed 2000 healthy adults using the Infinium global screening array (GSA) for 15 pharmacogenetic determinants of warfarin. In addition, we have elucidated the impact of these variants on protein function, stability, dynamics, evolutionary preservation, and ligand binding propensity. RESULTS The GSA Analysis has revealed that CYP4F2 V433M (MAF: 39.425%), VKORC1 -1639 G > A (MAF: 20.5%), CYP2C9*3 (MAF:9.925%), and CYP2C9*2 (MAF:4.575%) are common, while CYP2C9*14 (MAF: 1.475%), CYP2C9*4 (0.175%), CYP2C9*5 (0.125%), and CYP2C9*11 (0.125%) are rare. Position-specific evolutionary preservation (PSEP) analysis has revealed that CYP2C9*4 is possibly damaging, while CYP2C9*5, CYP2C9*11, and CYP2C9*14 are probably damaging. CYP2C9*4 has high thermolability (-10.14 kcal/mol). Among the rare CYP2C9 variants, CYP2C9*4 and CYP2C9*11 exert destabilizing effects and may have increased molecular flexibility, while CYP2C9*5 and CYP2C9*14 exert stabilizing effects and may have decreased molecular flexibility. DNase I footprint analysis has revealed the loss of the E-box consensus sequence due to VKORC1 -1639 G > A polymorphism. CONCLUSION CYP2C9*2, CYP2C9*3, VKORC1 -1639 G > A and CYP4F2 V433M are common; CYP2C9*4, CYP2C9*5, CYP2C9*11, and CYP2C9*14 variants are rare in Indian subjects. All the CYP2C9 variants are found to be damaging. DNase I footprint analysis provided the mechanistic rationale for the association of VKORC1 -1639 G > A with warfarin sensitivity.
Collapse
Affiliation(s)
- Shaik Mohammad Naushad
- Department of Biochemical Genetics and Pharmacogenomics, Sandor Speciality Diagnostics Pvt Ltd, Banjara Hills, Road No 3, Hyderabad, 500034, India.
| | - Vijay Kumar Kutala
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences, Hyderabad, India
| | - Tajamul Hussain
- Center of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Salman A Alrokayan
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
- Biochemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
25
|
Russell LE, Zhou Y, Almousa AA, Sodhi JK, Nwabufo CK, Lauschke VM. Pharmacogenomics in the era of next generation sequencing - from byte to bedside. Drug Metab Rev 2021; 53:253-278. [PMID: 33820459 DOI: 10.1080/03602532.2021.1909613] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pharmacogenetic research has resulted in the identification of a multitude of genetic variants that impact drug response or toxicity. These polymorphisms are mostly common and have been included as actionable information in the labels of numerous drugs. In addition to common variants, recent advances in Next Generation Sequencing (NGS) technologies have resulted in the identification of a plethora of rare and population-specific pharmacogenetic variations with unclear functional consequences that are not accessible by conventional forward genetics strategies. In this review, we discuss how comprehensive sequencing information can be translated into personalized pharmacogenomic advice in the age of NGS. Specifically, we provide an update of the functional impacts of rare pharmacogenetic variability and how this information can be leveraged to improve pharmacogenetic guidance. Furthermore, we critically discuss the current status of implementation of pharmacogenetic testing across drug development and layers of care. We identify major gaps and provide perspectives on how these can be minimized to optimize the utilization of NGS data for personalized clinical decision-support.
Collapse
Affiliation(s)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ahmed A Almousa
- Department of Pharmacy, London Health Sciences Center, Victoria Hospital, London, ON, Canada
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA.,Department of Drug Metabolism and Pharmacokinetics, Plexxikon, Inc., Berkeley, CA, USA
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Luo S, Jiang R, Grzymski JJ, Lee W, Lu JT, Washington NL. Comprehensive Allele Genotyping in Critical Pharmacogenes Reduces Residual Clinical Risk in Diverse Populations. Clin Pharmacol Ther 2021; 110:759-767. [PMID: 33930192 PMCID: PMC8453755 DOI: 10.1002/cpt.2279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/19/2021] [Indexed: 11/25/2022]
Abstract
Genomic‐guided pharmaceutical prescribing is increasingly recognized as an important clinical application of genetics. Accurate genotyping of pharmacogenomic (PGx) genes can be difficult, owing to their complex genetic architecture involving combinations of single‐nucleotide polymorphisms and structural variation. Here, we introduce the Helix PGx database, an open‐source star allele, genotype, and resulting metabolic phenotype frequency database for CYP2C9, CYP2C19, CYP2D6, and CYP4F2, based on short‐read sequencing of >86,000 unrelated individuals enrolled in the Helix DNA Discovery Project. The database is annotated using a pipeline that is clinically validated against a broad range of alleles and designed to call CYP2D6 structural variants with high (98%) accuracy. We find that CYP2D6 has greater allelic diversity than the other genes, manifest in both a long tail of low‐frequency star alleles, as well as a disproportionate fraction (36%) of all novel predicted loss‐of‐function variants identified. Across genes, we observe that many rare alleles (<0.1% frequency) in the overall cohort have 10 times higher frequency in one or more subgroups with non‐European genetic ancestry. Extending these PGx genotypes to predicted metabolic phenotypes, we demonstrate that >90% of the cohort harbors a high‐risk variant in one of the four pharmacogenes. Based on the recorded prescriptions for >30,000 individuals in the Healthy Nevada Project, combined with predicted PGx metabolic phenotypes, we anticipate that standard‐of‐care screening of these 4 pharmacogenes could impact nearly half of the general population.
Collapse
Affiliation(s)
| | | | - Joseph J Grzymski
- Desert Research Institute, Reno, Nevada, USA.,Renown Institute of Health Innovation, Reno, Nevada, USA
| | | | | | | |
Collapse
|
27
|
Díaz-Ordóñez L, Ramírez-Montaño D, Candelo E, González-Restrepo C, Silva-Peña S, Rojas CA, Sepulveda Copete M, Echavarria HR, Pachajoa H. Evaluation of CYP2C19 Gene Polymorphisms in Patients with Acid Peptic Disorders Treated with Esomeprazole. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:509-520. [PMID: 33953602 PMCID: PMC8092628 DOI: 10.2147/pgpm.s285144] [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: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 11/23/2022]
Abstract
Background CYP2C19 is a highly polymorphic gene that encodes an enzyme with the same name and whose function is associated with the metabolism of many important drugs, such as proton pump inhibitors (such as esomeprazole, which is used for the treatment of acid peptic disease). Genetic variants in CYP2C19 alter protein function and affect drug metabolism. This study aims to genotypically and phenotypically characterize the genetic variants in the CYP2C19 gene in 12 patients with acid peptic disorders and different therapeutic profiles to proton pump inhibitor (PPI) drugs. The patients were randomly selected from a controlled, randomized and blinded clinical pilot trial of 33 patients. We determined the presence and frequency of single nucleotide polymorphisms (SNPs) within exons 1–5 and 9, the intron-exon junctions, and a fragment in the 3ʹ UTR region of the CYP2C19 gene using Sanger sequencing. Undescribed polymorphisms were analyzed by free online bioinformatics tools to evaluate the potential molecular effects of these genetic variants. Results We identified nine polymorphisms, six of which had no reported functions. One of these genetic variants, with a functional impact, not yet reported (p.Arg132Trp) was predicted by bioinformatic tools as potentially pathogenic. This finding suggests that p.Arg132Trp could be related to poor metabolizers of drugs metabolized by CYP2C19. Conclusion We identified the genotype spectrum of variants in CYP2C19. The genotype spectrum of variants in CYP2C19 could predict the treatment response and could support to evaluate clinical efficacy in patients treated with esomeprazole.
Collapse
Affiliation(s)
- Lorena Díaz-Ordóñez
- Basic Medical Science Department, Faculty of Health Sciences, Universidad Icesi, Cali, Colombia.,Clinical Genetic Department, Fundación Valle del Lili, Cali, Colombia.,Research Centre in Rare Diseases and Congenital Abnormalities (CIACER), Universidad Icesi, Cali, Colombia
| | - Diana Ramírez-Montaño
- Basic Medical Science Department, Faculty of Health Sciences, Universidad Icesi, Cali, Colombia.,Clinical Genetic Department, Fundación Valle del Lili, Cali, Colombia.,Research Centre in Rare Diseases and Congenital Abnormalities (CIACER), Universidad Icesi, Cali, Colombia
| | - Estephania Candelo
- Clinical Genetic Department, Fundación Valle del Lili, Cali, Colombia.,Research Centre in Rare Diseases and Congenital Abnormalities (CIACER), Universidad Icesi, Cali, Colombia.,Research Centre, Fundación Valle de Lili, Cali, Colombia
| | | | - Sebastián Silva-Peña
- Basic Medical Science Department, Faculty of Health Sciences, Universidad Icesi, Cali, Colombia
| | | | | | | | - Harry Pachajoa
- Basic Medical Science Department, Faculty of Health Sciences, Universidad Icesi, Cali, Colombia.,Clinical Genetic Department, Fundación Valle del Lili, Cali, Colombia.,Research Centre in Rare Diseases and Congenital Abnormalities (CIACER), Universidad Icesi, Cali, Colombia
| |
Collapse
|
28
|
Zhang L, Sarangi V, Ho MF, Moon I, Kalari KR, Wang L, Weinshilboum RM. SLCO1B1: Application and Limitations of Deep Mutational Scanning for Genomic Missense Variant Function. Drug Metab Dispos 2021; 49:395-404. [PMID: 33658230 PMCID: PMC8042483 DOI: 10.1124/dmd.120.000264] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/17/2021] [Indexed: 01/07/2023] Open
Abstract
SLCO1B1 (solute carrier organic anion transporter family member 1B1) is an important transmembrane hepatic uptake transporter. Genetic variants in the SLCO1B1 gene have been associated with altered protein folding, resulting in protein degradation and decreased transporter activity. Next-generation sequencing (NGS) of pharmacogenes is being applied increasingly to associate variation in drug response with genetic sequence variants. However, it is difficult to link variants of unknown significance with functional phenotypes using "one-at-a-time" functional systems. Deep mutational scanning (DMS) using a "landing pad cell-based system" is a high-throughput technique designed to analyze hundreds of gene open reading frame (ORF) missense variants in a parallel and scalable fashion. We have applied DMS to analyze 137 missense variants in the SLCO1B1 ORF obtained from the Exome Aggregation Consortium project. ORFs containing these variants were fused to green fluorescent protein and were integrated into "landing pad" cells. Florescence-activated cell sorting was performed to separate the cells into four groups based on fluorescence readout indicating protein expression at the single cell level. NGS was then performed and SLCO1B1 variant frequencies were used to determine protein abundance. We found that six variants not previously characterized functionally displayed less than 25% and another 12 displayed approximately 50% of wild-type protein expression. These results were then functionally validated by transporter studies. Severely damaging variants identified by DMS may have clinical relevance for SLCO1B1-dependent drug transport, but we need to exercise caution since the relatively small number of severely damaging variants identified raise questions with regard to the application of DMS to intrinsic membrane proteins such as organic anion transporter protein 1B1. SIGNIFICANCE STATEMENT: The functional implications of a large numbers of open reading frame (ORF) "variants of unknown significance" (VUS) in transporter genes have not been characterized. This study applied deep mutational scanning to determine the functional effects of VUS that have been observed in the ORF of SLCO1B1(s olute carrier organic anion transporter family member 1B1). Several severely damaging variants were identified, studied, and validated. These observations have implications for both the application of deep mutational scanning to intrinsic membrane proteins and for the clinical effect of drugs and endogenous compounds transported by SLCO1B1.
Collapse
Affiliation(s)
- Lingxin Zhang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Vivekananda Sarangi
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Irene Moon
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Krishna R Kalari
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|