1
|
Borbón A, Briceño JC, Valderrama-Aguirre A. Pharmacogenomics Tools for Precision Public Health and Lessons for Low- and Middle-Income Countries: A Scoping Review. Pharmgenomics Pers Med 2025; 18:19-34. [PMID: 39902237 PMCID: PMC11789506 DOI: 10.2147/pgpm.s490135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/21/2024] [Indexed: 02/05/2025] Open
Abstract
Pharmacogenomics is the integration of genomics and pharmacology to optimize drug response and reduce side effects. In terms of personalized or individualized medicine, PGx is defined as the identification and analysis of specific genetic variants associated with particular drug treatments for each patient. Under a precision public health (PPH) approach, population-level data are analyzed to generate public health strategies. The objective of this study was to conduct a scoping review of technological tools, examining their evolution, the predominance of high-income countries in their development, and the gaps and needs for genomic data and advances in low- and middle-income countries (LMICs). This review was conducted in accordance with the ScPRISMA guidelines. A search was conducted in PubMed, Web of Science and Embase until January 2024. A total of 40 documents were selected, which revealed the continuous evolution and progressive development of pharmacogenomic tools. The technological tools developed come from high-income countries, particularly the United States, Canada, China, and several European nations, where international collaboration has been essential to maintain and expand these tools, which have evolved to keep pace with the rapid generation of genomic data. This trend shows a scarce development of technological tools for public health precision in LMICs, which evidences the need to increase investment in genomic research infrastructure in this aspect and in the development of capacities to guarantee global accessibility and boost PPH for all populations.
Collapse
Affiliation(s)
- Angélica Borbón
- Technological Innovation Management, University of the Andes, Bogotá, Colombia
| | - Juan Carlos Briceño
- Department of Biomedical Engineering, Director of Technological Innovation Management Programs, University of the Andes, Bogotá, Colombia
| | - Augusto Valderrama-Aguirre
- Department of Biological Sciences, Faculty of Sciences, Director of the Biomedical Research Institute Group, University of the Andes, Bogotá, Colombia
| |
Collapse
|
2
|
Dafniet B, Taboureau O. Prediction of adverse drug reactions due to genetic predisposition using deep neural networks. Mol Inform 2024; 43:e202400021. [PMID: 38850150 DOI: 10.1002/minf.202400021] [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: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 06/10/2024]
Abstract
Drug development is a long and costly process, often limited by the toxicity and adverse drug reactions (ADRs) caused by drug candidates. Even on the market, some drugs can cause strong ADRs that can vary depending on an individual polymorphism. The development of Genome-wide association studies (GWAS) allowed the discovery of genetic variants of interest that may cause these effects. In this study, the objective was to investigate a deep learning approach to predict genetic variations potentially related to ADRs. We used single nucleotide polymorphisms (SNPs) information from dbSNP to create a network based on ADR-drug-target-mutations and extracted matrixes of interaction to build deep Neural Networks (DNN) models. Considering only information about mutations known to impact drug efficacy and drug safety from PharmGKB and drug adverse reactions based on the MedDRA System Organ Classes (SOCs), these DNN models reached a balanced accuracy of 0.61 in average. Including molecular fingerprints representing structural features of the drugs did not improve the performance of the models. To our knowledge, this is the first model that exploits DNN to predict ADR-drug-target-mutations. Although some improvements are suggested, these models can be of interest to analyze multiple compounds over all of the genes and polymorphisms information accessible and thus pave the way in precision medicine.
Collapse
Affiliation(s)
- Bryan Dafniet
- INSERM U1133, CNRS UMR 8251, Université Paris Cité, 35 rue Hélène Brion, Paris, 75013, France
| | - Olivier Taboureau
- INSERM U1133, CNRS UMR 8251, Université Paris Cité, 35 rue Hélène Brion, Paris, 75013, France
| |
Collapse
|
3
|
Hong DZ, Ong TCC, Timbadia DP, Tan HTA, Kwa ED, Chong WQ, Goh BC, Loh WS, Loh KS, Tan EC, Tay JK. Systematic Review and Meta-Analysis of the Influence of Genetic Variation on Ototoxicity in Platinum-Based Chemotherapy. Otolaryngol Head Neck Surg 2023; 168:1324-1337. [PMID: 36802061 DOI: 10.1002/ohn.222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 11/04/2022] [Accepted: 11/19/2022] [Indexed: 02/19/2023]
Abstract
OBJECTIVE The objective of this meta-analysis is to evaluate the impact of genetic polymorphisms on platinum-based chemotherapy (PBC)-induced ototoxicity. DATA SOURCES Systematic searches of PubMed, Embase, Cochrane, and Web of Science were conducted from the inception of the databases to May 31, 2022. Abstracts and presentations from conferences were also reviewed. REVIEW METHODS Four investigators independently extracted data in adherence to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Differences in the prevalence of PBC-induced ototoxicity between reference and variant (i) genotypes and (ii) alleles were analyzed. The overall effect size was presented using the random-effects model as an odds ratio (OR) with a 95% confidence interval (CI). RESULTS From 32 included articles, 59 single nucleotide polymorphisms on 28 genes were identified, with 4406 total unique participants. For allele frequency analysis, the A allele in ACYP2 rs1872328 was positively associated with ototoxicity (OR: 2.61; 95% CI: 1.06-6.43; n = 2518). Upon limiting to cisplatin use only, the T allele of COMT rs4646316 and COMT rs9332377 revealed significant results. For genotype frequency analysis, the CT/TT genotype in ERCC2 rs1799793 demonstrated an otoprotective effect (OR: 0.50; 95% CI: 0.27-0.94; n = 176). Excluding studies using carboplatin or concomitant radiotherapy revealed significant effects with COMT rs4646316, GSTP1 rs1965, and XPC rs2228001. Major sources of variations between studies include differences in patient demographics, ototoxicity grading systems, and treatment protocols. CONCLUSION Our meta-analysis presents polymorphisms that exert ototoxic or otoprotective effects in patients undergoing PBC. Importantly, several of these alleles are observed at high frequencies globally, highlighting the potential for polygenic screening and cumulative risk evaluation for personalized care.
Collapse
Affiliation(s)
- Daniel Z Hong
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Thaned C C Ong
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Dhayan P Timbadia
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Hui T A Tan
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
| | - Eunice D Kwa
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Wan Q Chong
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Boon C Goh
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Woei S Loh
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Kwok S Loh
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| | - Ene C Tan
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Joshua K Tay
- Department of Otolaryngology-Head and Neck Surgery, National University of Singapore, Singapore, Singapore
- Department of Otolaryngology-Head and Neck Surgery, National University Hospital, Singapore, Singapore
| |
Collapse
|
4
|
Roosan D, Chok J, Baskys A, Roosan MR. PGxKnow: a pharmacogenomics educational HoloLens application of augmented reality and artificial intelligence. Pharmacogenomics 2022; 23:235-245. [PMID: 35083917 DOI: 10.2217/pgs-2021-0120] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop and assess an augmented reality tool for pharmacogenomics (PGx) education based on artificial intelligence. Materials & methods: A HoloLens application was developed using feedback from three clinical PGx-trained pharmacists. 15 Participants independently reviewed the application and assessed usability using the system usability scale (SUS). Results & conclusion: Eighteen different frames were developed. Each video module was 2-3 min for the education. The application included textual information and 3D structures of PGx concepts. The mean SUS score for 15 participants (11 pharmacy students and four pharmacists) was 83, with a standard deviation of 6.6. Results suggest that PGxKnow has the potential to bridge the gap in PGx education, further widespread utilization of PGx and boost its impact on precision medicine.
Collapse
Affiliation(s)
- Don Roosan
- Assistant Professor, College of Pharmacy, Department of Pharmacy Practice and Administration, Western University of Health Sciences, 309 E 2nd street, Pomona, CA 91766, USA
| | - Jay Chok
- Associate Professor, Claremont Colleges, School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Andrius Baskys
- Professor, College of Graduate Biomedical Sciences, Western University of Health Sciences, 309 E 2nd street, Pomona, CA 91766, USA
| | - Moom R Roosan
- Assistant Professor, School of Pharmacy, Department of Pharmacy Practice, Chapman University, 9401 Jeronimo Road, Irvine, CA 92618, USA
| |
Collapse
|
5
|
Piriyapongsa J, Sukritha C, Kaewprommal P, Intarat C, Triparn K, Phornsiricharoenphant K, Chaosrikul C, Shaw PJ, Chantratita W, Mahasirimongkol S, Tongsima S. PharmVIP: A Web-Based Tool for Pharmacogenomic Variant Analysis and Interpretation. J Pers Med 2021; 11:1230. [PMID: 34834582 PMCID: PMC8618518 DOI: 10.3390/jpm11111230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/17/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
The increasing availability of next generation sequencing (NGS) for personal genomics could promote pharmacogenomics (PGx) discovery and application. However, current tools for analysis and interpretation of pharmacogenomic variants from NGS data are inadequate, as none offer comprehensive analytic functions in a simple, web-based platform. In addition, no tools exist to analyze human leukocyte antigen (HLA) genes for determining potential risks of immune-mediated adverse drug reaction (IM-ADR). We describe PharmVIP, a web-based PGx tool, for one-stop comprehensive analysis and interpretation of genome-wide variants obtained from NGS platforms. PharmVIP comprises three main interpretation modules covering analyses of pharmacogenes involved in pharmacokinetics, pharmacodynamics and IM-ADR. The Guideline module provides Clinical Pharmacogenetics Implementation Consortium (CPIC) drug guideline recommendations based on the translation of genotypic data in genes having guidelines. The HLA module reports HLA genotypes, potential adverse drug reactions, and the relevant drug guidelines. The Pharmacogenes module is employed for prioritizing variants according to variant effect on gene function. Detailed, customizable reports are provided as exportable files and as an interactive web version. PharmVIP is a new integrated NGS workflow for the PGx community to facilitate discovery and clinical application.
Collapse
Affiliation(s)
- Jittima Piriyapongsa
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Chanathip Sukritha
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Pavita Kaewprommal
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Chalermpong Intarat
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Kwankom Triparn
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Krittin Phornsiricharoenphant
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Chadapohn Chaosrikul
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| | - Philip J. Shaw
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand;
| | - Wasun Chantratita
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Phayathai, Bangkok 10400, Thailand;
| | - Surakameth Mahasirimongkol
- Division of Genomic Medicine and Innovation Support, Department of Medical Sciences, Ministry of Public Health, Nonthaburi 11000, Thailand;
| | - Sissades Tongsima
- National Biobank of Thailand, National Science and Technology Development Agency, Klong Luang, Pathum Thani 12120, Thailand; (C.S.); (P.K.); (C.I.); (K.T.); (K.P.); (C.C.); (S.T.)
| |
Collapse
|
6
|
Pharmacogenes (PGx-genes): Current understanding and future directions. Gene 2019; 718:144050. [DOI: 10.1016/j.gene.2019.144050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022]
|
7
|
Sabater A, Ciudad CJ, Cendros M, Dobrokhotov D, Sabater-Tobella J. g-Nomic: a new pharmacogenetics interpretation software. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:75-85. [PMID: 31239753 PMCID: PMC6554524 DOI: 10.2147/pgpm.s203585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/27/2019] [Indexed: 11/23/2022]
Abstract
We present g-Nomic, a pharmacogenetics interpretation software that analyzes globally a prescribed medication taking into account the personal background genetics, drug–drug interactions, lifestyle, nutritional supplements, inhibitors, inducers, and other risks to analyze primary or secondary metabolism pathways. G-Nomic provides a set of recommendations describing the suitability of a given combination of drugs for each patient according to their genes and polymedication. G-Nomic is updated monthly including data from the new drugs to be included, their known interactions, and the relevant pharmacokinetic biomarkers. For the interactions, the list is curated manually, only keeping those with clinical relevance. For each drug, their FDA and EMA drug labels are accessed, to check for relevant enzymes and transport proteins that influence its pharmacokinetics, and for their ability to induce or inhibit other enzymes, particularly the CYP-450 system. When this information is not available, a PubMed search is made to look for these characteristics. In addition, a distinction is made between drugs and prodrugs. A query on the g-Nomic software begins with entering the medication by either their common or commercial name. Non-pharmacological substances can be also added or selected under “lifestyle habits”. The lifestyle list is dynamic, showing only the substances known to interact with the drugs that are currently selected, and includes herb compounds, such as St. John’s wort, as well as proper lifestyle substances such as grapefruit or cigarette smoking. The software provides a list of the genes classified as primary biomarkers as candidates for genetic testing, and a list of the interactions that have been detected. If genetic information is available then, or is made available at a later point, these results can also be entered and the software returns pharmacogenetics recommendations regarding specific genotypes. g-Nomic takes all the above-mentioned parameters in an easy and user-friendly tool making prescription safer.
Collapse
Affiliation(s)
- Ana Sabater
- Department of Information Technology, EUGENOMIC, Barcelona 08012, Spain
| | - Carlos J Ciudad
- Department of Biochemistry and Physiology, University of Barcelona, Barcelona 08028, Spain
| | - Marc Cendros
- Department of Information Technology, EUGENOMIC, Barcelona 08012, Spain
| | - Denis Dobrokhotov
- Department of Information Technology, EUGENOMIC, Barcelona 08012, Spain
| | | |
Collapse
|
8
|
Sivadas A, Scaria V. Population-scale genomics-Enabling precision public health. ADVANCES IN GENETICS 2018; 103:119-161. [PMID: 30904093 DOI: 10.1016/bs.adgen.2018.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The current excitement for affordable genomics technologies and national precision medicine initiatives marks a turning point in worldwide healthcare practices. The last decade of global population sequencing efforts has defined the enormous extent of genetic variation in the human population resulting in insights into differential disease burden and response to therapy within and between populations. Population-scale pharmacogenomics helps to provide insights into the choice of optimal therapies and an opportunity to estimate, predict and minimize adverse events. Such an approach can potentially empower countries to formulate national selection and dosing policies for therapeutic agents thereby promoting public health with precision. We review the breadth and depth of worldwide population-scale sequencing efforts and its implications for the implementation of clinical pharmacogenetics toward making precision medicine a reality.
Collapse
Affiliation(s)
- Ambily Sivadas
- GN Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Vinod Scaria
- GN Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
9
|
Drögemöller BI, Brooks B, Critchley C, Monzon JG, Wright GEB, Liu G, Renouf DJ, Kollmannsberger CK, Bedard PL, Hayden MR, Gelmon KA, Carleton BC, Ross CJD. Further Investigation of the Role of ACYP2 and WFS1 Pharmacogenomic Variants in the Development of Cisplatin-Induced Ototoxicity in Testicular Cancer Patients. Clin Cancer Res 2018; 24:1866-1871. [PMID: 29358504 DOI: 10.1158/1078-0432.ccr-17-2810] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/01/2017] [Accepted: 01/16/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Adverse drug reactions such as ototoxicity, which occurs in approximately one-fifth of adult patients who receive cisplatin treatment, can incur large socioeconomic burdens on patients with testicular cancer who develop this cancer during early adulthood. Recent genome-wide association studies have identified genetic variants in ACYP2 and WFS1 that are associated with cisplatin-induced ototoxicity. We sought to explore the role of these genetic susceptibility factors to cisplatin-induced ototoxicity in patients with testicular cancer.Experimental Design: Extensive clinical and demographic data were collected for 229 patients with testicular cancer treated with cisplatin. Patients were genotyped for two variants, ACYP2 rs1872328 and WFS1 rs62283056, that have previously been associated with hearing loss in cisplatin-treated patients. Analyses were performed to investigate the association of these variants with ototoxicity in this cohort of adult patients with testicular cancer.Results: Pharmacogenomic analyses revealed that ACYP2 rs1872328 was significantly associated with cisplatin-induced ototoxicity [P = 2.83 × 10-3, OR (95% CI):14.7 (2.6-84.2)]. WFS1 rs62283056 was not significantly associated with ototoxicity caused by cisplatin (P = 0.39); however, this variant was associated with hearing loss attributable to any cause [P = 5.67 × 10-3, OR (95% CI): 3.2 (1.4-7.7)].Conclusions: This study has provided the first evidence for the role of ACYP2 rs1872328 in cisplatin-induced ototoxicity in patients with testicular cancer. These results support the use of this information to guide the development of strategies to prevent cisplatin-induced ototoxicity across cancers. Further, this study has highlighted the importance of phenotypic differences in replication studies and has provided further evidence for the role of WFS1 rs62283056 in susceptibility to hearing loss, which may be worsened by cisplatin treatment. Clin Cancer Res; 24(8); 1866-71. ©2018 AACR.
Collapse
Affiliation(s)
- Britt I Drögemöller
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Beth Brooks
- Audiology and Speech Pathology Department, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Carol Critchley
- Neuro-Otology Unit, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | | | - Galen E B Wright
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Geoffrey Liu
- Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre-University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Daniel J Renouf
- BC Cancer Agency and University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Philippe L Bedard
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | - Michael R Hayden
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen A Gelmon
- BC Cancer Agency and University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce C Carleton
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada
| | - Colin J D Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada. .,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Gonzalez-Covarrubias V, Urena-Carrion J, Villegas-Torres B, Cossío-Aranda JE, Trevethan-Cravioto S, Izaguirre-Avila R, Fiscal-López OJ, Soberon X. Pharmacogenetic Variation in Over 100 Genes in Patients Receiving Acenocumarol. Front Pharmacol 2017; 8:863. [PMID: 29218011 PMCID: PMC5703746 DOI: 10.3389/fphar.2017.00863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/09/2017] [Indexed: 12/18/2022] Open
Abstract
Coumarins are widely prescribed worldwide, and in Mexico acenocumarol is the preferred form. It is well known that despite its efficacy, coumarins show a high variability for dose requirements. We investigated the pharmacogenetic variation of 110 genes in patients receiving acenocumarol using a targeted NGS approach. We report relevant population differentiation for variants on CYP2C8, CYP2C19, CYP4F11, CYP4F2, PROS, and GGCX, VKORC1, CYP2C18, NQO1. A higher proportion of novel-to-known variants for 10 genes was identified on 41 core pharmacogenomics genes related to the PK (29), PD (3), of coumarins, and coagulation proteins (9) including, CYP1A1, CYP3A4, CYP3A5, and F8, and a low proportion of novel-to-known variants on CYP2E1, VKORC1, and SULT1A1/2. Using a Bayesian approach, we identified variants influencing acenocumarol dosing on, VKORC1 (2), SULT1A1 (1), and CYP2D8P (1) explaining 40–55% of dose variability. A collection of pharmacogenetic variation on 110 genes related to the PK/PD of coumarins is also presented. Our results offer an initial insight into the use of a targeted NGS approach in the pharmacogenomics of coumarins in Mexican Mestizos.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xavier Soberon
- Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| |
Collapse
|
11
|
ePGA: A Web-Based Information System for Translational Pharmacogenomics. PLoS One 2016; 11:e0162801. [PMID: 27631363 PMCID: PMC5025168 DOI: 10.1371/journal.pone.0162801] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/29/2016] [Indexed: 11/19/2022] Open
Abstract
One of the challenges that arise from the advent of personal genomics services is to efficiently couple individual data with state of the art Pharmacogenomics (PGx) knowledge. Existing services are limited to either providing static views of PGx variants or applying a simplistic match between individual genotypes and existing PGx variants. Moreover, there is a considerable amount of haplotype variation associated with drug metabolism that is currently insufficiently addressed. Here, we present a web-based electronic Pharmacogenomics Assistant (ePGA; http://www.epga.gr/) that provides personalized genotype-to-phenotype translation, linked to state of the art clinical guidelines. ePGA's translation service matches individual genotype-profiles with PGx gene haplotypes and infers the corresponding diplotype and phenotype profiles, accompanied with summary statistics. Additional features include i) the ability to customize translation based on subsets of variants of clinical interest, and ii) to update the knowledge base with novel PGx findings. We demonstrate ePGA's functionality on genetic variation data from the 1000 Genomes Project.
Collapse
|