1
|
Zheng Y, Tang M, Deng Z, Cai P. Genetic polymorphisms and platinum-induced hematological toxicity: a systematic review. Front Pharmacol 2024; 15:1445328. [PMID: 39234108 PMCID: PMC11371761 DOI: 10.3389/fphar.2024.1445328] [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: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Background Platinum-based chemotherapy bring severe hematological toxicity that can lead to dose reduction or discontinuation of therapy. Genetic variations have been reported to influence the risk and extent of hematological toxicity; however, the results are controversial and a comprehensive overview is lacking. This systematic review aimed to identify genetic biomarkers of platinum-induced hematological toxicity. Method Pubmed, Embase and Web of science database were systematically reviewed for studies that evaluated the association of genetic variants and platinum-related hematological toxicity in tumor patients with no prior history of chemotherapy or radiation, published from inception to the 28th of January 2022. The studies should have specific toxicity scoring system as well as defined toxicity end-point. The quality of reporting was assessed using the Strengthening the Reporting of Genetic Association Studies (STREGA) checklist. Results were summarized using narrative synthesis. Results 83 studies were eligible with over 682 single-nucleotide polymorphisms across 110 genes. The results are inconsistent and diverse with methodological issues including insufficient sample size, population stratification, various treatment schedule and toxicity end-point, and inappropriate statistics. 11 SNPs from 10 genes (ABCB1 rs1128503, GSTP1 rs1695, GSTM1 gene deletion, ERCC1 rs11615, ERCC1 rs3212986, ERCC2 rs238406, XPC rs2228001, XPCC1 rs25487, MTHFR rs1801133, MDM2 rs2279744, TP53 rs1042522) had consistent results in more than two independent populations. Among them, GSTP1 rs1695, ERCC1 rs11615, ERCC1 rs3212986, and XRCC1 rs25487 present the most promising results. Conclusion Even though the results are inconsistent and several methodological concerns exist, this systematic review identified several genetic variations that deserve validation in well-defined studies with larger sample size and robust methodology. Systematic Review Registration https://www.crd.york.ac.uk/, identifier CRD42021234164.
Collapse
Affiliation(s)
- Yi Zheng
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Deng
- Hunan Institute for Tuberculosis Control and Hunan Chest Hospital, Changsha, China
- Hunan Chest Hospital, Changsha, China
| | - Pei Cai
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| |
Collapse
|
2
|
Lusiki Z, Blom D, Soko ND, Malema S, Jones E, Rayner B, Blackburn J, Sinxadi P, Dandara MT, Dandara C. Major Genetic Drivers of Statin Treatment Response in African Populations and Pharmacogenetics of Dyslipidemia Through a One Health Lens. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:261-279. [PMID: 37956269 DOI: 10.1089/omi.2023.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
A One Health lens is increasingly significant to address the intertwined challenges in planetary health concerned with the health of humans, nonhuman animals, plants, and ecosystems. A One Health approach can benefit the public health systems in Africa that are overburdened by noncommunicable, infectious, and environmental diseases. Notably, the COVID-19 pandemic revealed the previously overlooked two-fold importance of pharmacogenetics (PGx), for individually tailored treatment of noncommunicable diseases and environmental pathogens. For example, dyslipidemia, a common cardiometabolic risk factor, has been identified as an independent COVID-19 severity risk factor. Observational data suggest that patients with COVID-19 infection receiving lipid-lowering therapy may have better outcomes. However, among African patients, the response to these drugs varies from patient to patient, pointing to the possible contribution of genetic variation in important pharmacogenes. The PGx of lipid-lowering therapies may underlie differences in treatment responses observed among dyslipidemia patients as well as patients comorbid with COVID-19 and dyslipidemia. Genetic variations in APOE, ABCB1, CETP, CYP2C9, CYP3A4, CYP3A5, HMGCR, LDLR, NPC1L1, and SLCO1B1 genes affect the pharmacogenomics of statins, and they have individually been linked to differential responses to dyslipidemia and COVID-19 treatment. African populations are underrepresented in PGx research. This leads to poor accounting of additional diverse genetic variants that could be important in understanding interindividual and between-population variations in therapeutic responses to dyslipidemia and COVID-19. This expert review examines and synthesizes the salient and priority PGx variations, as seen through a One Health lens in Africa, to improve and inform personalized medicine in both dyslipidemia and COVID-19.
Collapse
Affiliation(s)
- Zizo Lusiki
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Dirk Blom
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Nyarai D Soko
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Smangele Malema
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Erika Jones
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Nephrology and Hypertension, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Brian Rayner
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Nephrology and Hypertension, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Phumla Sinxadi
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Michelle T Dandara
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| |
Collapse
|
3
|
Dua P, Seth S, Prasher B, Mukerji M, Maulik SK, Reeta KH. Pharmacogenomic biomarkers in coronary artery disease: a narrative review. Biomark Med 2024; 18:191-202. [PMID: 38456296 DOI: 10.2217/bmm-2023-0476] [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: 03/09/2024] Open
Abstract
Coronary artery disease (CAD) has a high mortality rate. Despite various therapeutic targets, non-responsiveness to drugs remains a prevalent issue. Pharmacogenomics assesses the way an individual's genetic attributes affect their likely response to drug therapy. Single-nucleotide polymorphisms play a crucial role in determining these outcomes. This review offers an overview of single-nucleotide polymorphisms investigated in clinical studies and their associations with drug response/nonresponse in the treatment of CAD. A total of 104 studies of whole sets of chromosomes and several genes were explored. A total of 161 polymorphisms exhibited associations with drug response/nonresponse in CAD across diverse ethnic populations. This pool can serve as a pharmacogenomic biomarker for predicting response to drug therapy in patients with CAD.
Collapse
Affiliation(s)
- Pamila Dua
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Sandeep Seth
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | - Mitali Mukerji
- Indian Institute of Technology, Jodhpur, Rajasthan, India
| | | | - K H Reeta
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| |
Collapse
|
4
|
Stewart S, Dodero-Anillo JM, Guijarro-Eguinoa J, Arias P, Gómez López De Las Huertas A, Seco-Meseguer E, García-García I, Ramírez García E, Rodríguez-Antolín C, Carcas AJ, Rodriguez-Novoa S, Rosas-Alonso R, Borobia AM. Advancing pharmacogenetic testing in a tertiary hospital: a retrospective analysis after 10 years of activity. Front Pharmacol 2023; 14:1292416. [PMID: 37927587 PMCID: PMC10622662 DOI: 10.3389/fphar.2023.1292416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
The field of pharmacogenetics (PGx) holds great promise in advancing personalized medicine by adapting treatments based on individual genetic profiles. Despite its benefits, there are still economic, ethical and institutional barriers that hinder its implementation in our healthcare environment. A retrospective analysis approach of anonymized data sourced from electronic health records was performed, encompassing a diverse patient population and evaluating key parameters such as prescribing patterns and test results, to assess the impact of pharmacogenetic testing. A head-to-head comparison with previously published activity results within the same pharmacogenetic laboratory was also conducted to contrast the progress made after 10 years. The analysis revealed significant utilization of pharmacogenetic testing in daily clinical practice, with 1,145 pharmacogenetic tests performed over a 1-year period and showing a 35% growth rate increase over time. Of the 17 different medical departments that sought PGx tests, the Oncology department accounted for the highest number, representing 58.47% of all genotyped patients. A total of 1,000 PGx tests were requested for individuals susceptible to receive a dose modification based on genotype, and 76 individuals received a genotype-guided dose adjustment. This study presents a comprehensive descriptive analysis of real-world data obtained from a public tertiary hospital laboratory specialized in pharmacogenetic testing, and presents data that strongly endorse the integration of pharmacogenetic testing into everyday clinical practice.
Collapse
Affiliation(s)
- Stefan Stewart
- Clinical Pharmacology Department, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | | | | | - Pedro Arias
- Pharmacogenetics Laboratory, Genetics Department, La Paz University Hospital, Madrid, Spain
| | | | | | - Irene García-García
- Clinical Pharmacology Department, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Elena Ramírez García
- Clinical Pharmacology Department, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Pharmacology Department, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Rodríguez-Antolín
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital La Paz Institute for Health Research—IdiPAZ, Madrid, Spain
| | - Antonio J. Carcas
- Clinical Pharmacology Department, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Pharmacology Department, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sonia Rodriguez-Novoa
- Genetics of Metabolic Diseases Laboratory, Genetics Department, La Paz University Hospital, Madrid, Spain
| | - Rocio Rosas-Alonso
- Pharmacogenetics Laboratory, Genetics Department, La Paz University Hospital, Madrid, Spain
- Experimental Therapies and Novel Biomarkers in Cancer, Hospital La Paz Institute for Health Research—IdiPAZ, Madrid, Spain
| | - Alberto M. Borobia
- Clinical Pharmacology Department, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Pharmacology Department, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
5
|
Xie Y, Zhai S, Jiang W, Zhao H, Mehrotra DV, Shen J. Statistical assessment of biomarker replicability using MAJAR method. Stat Methods Med Res 2023; 32:1961-1972. [PMID: 37519295 DOI: 10.1177/09622802231188519] [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: 08/01/2023]
Abstract
In the era of precision medicine, many biomarkers have been discovered to be associated with drug efficacy and safety responses, which can be used for patient stratification and drug response prediction. Due to the small sample size and limited power of randomized clinical studies, meta-analysis is usually conducted to aggregate all available studies to maximize the power for identifying prognostic and predictive biomarkers. However, it is often challenging to find an independent study to replicate the discoveries from the meta-analysis (e.g. meta-analysis of pharmacogenomics genome-wide association studies (PGx GWAS)), which seriously limits the potential impacts of the discovered biomarkers. To overcome this challenge, we develop a novel statistical framework, MAJAR (meta-analysis of joint effect associations for biomarker replicability assessment), to jointly test prognostic and predictive effects and assess the replicability of identified biomarkers by implementing an enhanced expectation-maximization algorithm and calculating their posterior-probability-of-replicabilities and Bayesian false discovery rates (Fdr). Extensive simulation studies were conducted to compare the performance of MAJAR and existing methods in terms of Fdr, power, and computational efficiency. The simulation results showed improved statistical power with well-controlled Fdr of MAJAR over existing methods and robustness to outliers under different data generation processes. We further demonstrated the advantages of MAJAR over existing methods by applying MAJAR to the PGx GWAS summary statistics data from a large cardiovascular randomized clinical trial. Compared to testing main effects only, MAJAR identified 12 novel variants associated with the treatment-related low-density lipoprotein cholesterol reduction from baseline.
Collapse
Affiliation(s)
- Yuhan Xie
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Song Zhai
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, USA
| | - Wei Jiang
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., North Wales, PA, USA *These authors contributed equally to this work
| | - Judong Shen
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, USA
| |
Collapse
|
6
|
Jansen ME, Rigter T, Fleur TMC, Souverein PC, Verschuren WMM, Vijverberg SJ, Swen JJ, Rodenburg W, Cornel MC. Predictive Value of SLCO1B1 c.521T>C Polymorphism on Observed Changes in the Treatment of 1136 Statin-Users. Genes (Basel) 2023; 14:456. [PMID: 36833383 PMCID: PMC9957000 DOI: 10.3390/genes14020456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Pharmacogenomic testing is a method to prevent adverse drug reactions. Pharmacogenomics could be relevant to optimize statin treatment, by identifying patients at high risk for adverse drug reactions. We aim to investigate the clinical validity and utility of pre-emptive pharmacogenomics screening in primary care, with SLCO1B1 c.521T>C as a risk factor for statin-induced adverse drug reactions. The focus was on changes in therapy as a proxy for adverse drug reactions observed in statin-users in a population-based Dutch cohort. In total, 1136 statin users were retrospectively genotyped for the SLCO1B1 c.521T>C polymorphism (rs4149056) and information on their statin dispensing was evaluated as cross-sectional research. Approximately half of the included participants discontinued or switched their statin treatment within three years. In our analyses, we could not confirm an association between the SLCO1B1 c.521T>C genotype and any change in statin therapy or arriving at a stable dose sooner in primary care. To be able to evaluate the predictive values of SLCO1B1 c.521T>C genotype on adverse drug reactions from statins, prospective data collection of actual adverse drug reactions and reasons to change statin treatment should be facilitated.
Collapse
Affiliation(s)
- Marleen E. Jansen
- Department of Clinical Genetics, Amsterdam Public Health Research Institute, Personalized Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Section Community Genetics, 1081 HV Amsterdam, The Netherlands
- Centre for Health Protection, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Tessel Rigter
- Department of Clinical Genetics, Amsterdam Public Health Research Institute, Personalized Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Section Community Genetics, 1081 HV Amsterdam, The Netherlands
- Centre for Health Protection, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Thom M. C. Fleur
- Department of Clinical Genetics, Amsterdam Public Health Research Institute, Personalized Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Section Community Genetics, 1081 HV Amsterdam, The Netherlands
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Patrick C. Souverein
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - W. M. Monique Verschuren
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| | - Susanne J. Vijverberg
- Department of Pulmonary Medicine and Amsterdam Public Health Research Institute, Personalized Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jesse J. Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Wendy Rodenburg
- Centre for Health Protection, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Martina C. Cornel
- Department of Clinical Genetics, Amsterdam Public Health Research Institute, Personalized Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Section Community Genetics, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
7
|
Kim S, Seo JD, Yun YM, Kim H, Kim TE, Lee T, Lee TR, Lee JH, Cho EH, Ki CS. Pharmacokinetics and Genetic Factors of Atorvastatin in Healthy Korean Subjects. Front Genet 2022; 13:836970. [PMID: 35664336 PMCID: PMC9160745 DOI: 10.3389/fgene.2022.836970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Statins are the most popular agents for the primary and secondary prevention of cardiovascular disease; however, the pharmacokinetic parameters and associated genetic factors in the Korean population have not been fully elucidated. This study explored the pharmacokinetic properties of atorvastatin and the association between genetic variations and atorvastatin pharmacokinetics in healthy Korean subjects. Methods: Atorvastatin (80 mg) was administered to 35 healthy Korean volunteers. Plasma levels of atorvastatin and its metabolites were measured sequentially using liquid chromatography-tandem mass spectrometry from 0 to 24 h after atorvastatin administration. Customized next-generation sequencing analysis was performed covering all coding exons of 15 genes, as well as 46 single-nucleotide variants in 29 genes related to statin pharmacokinetics. Results: The mean area under the concentration-time (AUC) and Cmax (maximum peak concentration) were 269.0 ng/ml∙h and 84.3 ng/ml, respectively, which were approximately two times higher than those reported in Caucasians. Genetic analysis revealed that eight genetic variants in ABCB1, ABCG2, APOA5, CETP, and CYP7A1 contributed to the AUC of atorvastatin. The atorvastatin AUC0–24 h prediction model was developed based on age and eight genetic variants using multivariate linear regression (adjusted R2 = 0.878, p < 0.0001). Conclusion: This study shows that the pharmacokinetic properties of atorvastatin in Koreans are different from those in Caucasians and that atorvastatin AUC0–24 h could be predicted based on age and eight genetic variants of ABCB1, ABCG2, APOA5, CETP, and CYP7A1.
Collapse
Affiliation(s)
- Serim Kim
- Department of Laboratory Medicine, Shinwon Medical Foundation, Gwangmyeong-si, South Korea
| | - Jong Do Seo
- Department of Laboratory Medicine, Konkuk University Medical Center, Seoul, South Korea
| | - Yeo-Min Yun
- Department of Laboratory Medicine, Konkuk University Medical Center, Seoul, South Korea
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
- *Correspondence: Yeo-Min Yun,
| | - Hanah Kim
- Department of Laboratory Medicine, Konkuk University Medical Center, Seoul, South Korea
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Tae-Eun Kim
- Department of Clinical Pharmacology, Konkuk University Medical Center, Seoul, South Korea
| | | | | | - Jun Hyung Lee
- Department of Laboratory Medicine, GC Labs, Yongin-si, South Korea
| | | | | |
Collapse
|
8
|
Trompet S, Postmus I, Warren HR, Noordam R, Smit RAJ, Theusch E, Li X, Arsenault B, Chasman DI, Hitman GA, Munroe PB, Rotter JI, Psaty BM, Caulfield MJ, Krauss RM, Cupples AL, Jukema WJ. The Pharmacogenetics of Statin Therapy on Clinical Events: No Evidence that Genetic Variation Affects Statin Response on Myocardial Infarction. Front Pharmacol 2022; 12:679857. [PMID: 35069183 PMCID: PMC8769168 DOI: 10.3389/fphar.2021.679857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/01/2021] [Indexed: 01/02/2023] Open
Abstract
Background: The pharmacogenetic effect on cardiovascular disease reduction in response to statin treatment has only been assessed in small studies. In a pharmacogenetic genome wide association study (GWAS) analysis within the Genomic Investigation of Statin Therapy (GIST) consortium, we investigated whether genetic variation was associated with the response of statins on cardiovascular disease risk reduction. Methods: The investigated endpoint was incident myocardial infarction (MI) defined as coronary heart disease death and definite and suspect non-fatal MI. For imputed single nucleotide polymorphisms (SNPs), regression analysis was performed on expected allelic dosage and meta-analysed with a fixed-effects model, inverse variance weighted meta-analysis. All SNPs with p-values <5.0 × 10−4 in stage 1 GWAS meta-analysis were selected for further investigation in stage-2. As a secondary analysis, we extracted SNPs from the Stage-1 GWAS meta-analysis results based on predefined hypotheses to possibly modifying the effect of statin therapy on MI. Results: In stage-1 meta-analysis (eight studies, n = 10,769, 4,212 cases), we observed no genome-wide significant results (p < 5.0 × 10−8). A total of 144 genetic variants were followed-up in the second stage (three studies, n = 1,525, 180 cases). In the combined meta-analysis, no genome-wide significant hits were identified. Moreover, none of the look-ups of SNPs known to be associated with either CHD or with statin response to cholesterol levels reached Bonferroni level of significance within our stage-1 meta-analysis. Conclusion: This GWAS analysis did not provide evidence that genetic variation affects statin response on cardiovascular risk reduction. It does not appear likely that genetic testing for predicting effects of statins on clinical events will become a useful tool in clinical practice.
Collapse
Affiliation(s)
- Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Iris Postmus
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom.,Barts NIHR Biomedical Research Unit, London, United Kingdom
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands.,Department of Epidemiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Roelof A J Smit
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elizabeth Theusch
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Benoit Arsenault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec City, QC, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Graham A Hitman
- Blizard institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom.,Barts NIHR Biomedical Research Unit, London, United Kingdom
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States.,Department of Epidemiology, University of Washington, Seattle, WA, United States.,Department of Health Services University of Washington, Seattle, WA, United States
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom.,Barts NIHR Biomedical Research Unit, London, United Kingdom
| | - Ron M Krauss
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Adrienne L Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States.,NHLBI Framingham Heart Study, Framingham, MA, United States
| | - Wouter J Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| |
Collapse
|
9
|
Sheikhy A, Fallahzadeh A, Aghaei Meybodi HR, Hasanzad M, Tajdini M, Hosseini K. Personalized medicine in cardiovascular disease: review of literature. J Diabetes Metab Disord 2021; 20:1793-1805. [PMID: 34900826 DOI: 10.1007/s40200-021-00840-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
Purpose Personalized medicine (PM) is the concept of managing patients based on their characteristics, including genotypes. In the field of cardiology, advantages of PM could be found in the diagnosis and treatment of several conditions such as arrhythmias and cardiomyopathies; moreover, it may be beneficial to prevent adverse drug reactions (ADR) and select the best medication. Genetic background can help us in selecting effective treatments, appropriate dose requirements, and preventive strategies in individuals with particular genotypes. Method In this review, we provide examples of personalized medicine based on human genetics for the most used pharmaceutics in cardiology, including warfarin, clopidogrel, and statins. We also review cardiovascular diseases, including coronary artery disease, arrhythmia, and cardiomyopathies. Conclusion Genetic factors are as important as environmental factors and they should be tested and evaluated more in the future by improving in genetic testing tools. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-021-00840-0.
Collapse
Affiliation(s)
- Ali Sheikhy
- Research Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Fallahzadeh
- Research Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghaei Meybodi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Masih Tajdini
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kaveh Hosseini
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Niu RB, Dong XX, Guo LP, Pan L, Hai YQ, Chen XX, Duan BS. Study on the relationship between SLCO1B1 and ApoE gene polymorphisms and the risk of coronary heart disease in the Mongolian population. Clin Exp Hypertens 2021; 43:788-792. [PMID: 34423691 DOI: 10.1080/10641963.2021.1969660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objective This study aimed to analyze the influence of SLCO1B1 and APOE gene polymorphisms on coronary heart disease in Mongolian population who living in Ordos area. Methods From January 2019 to June 2020, 200 Mongolian patients with coronary heart disease admitted to our hospital and other banner hospitals were selected as the case group. At the same time, 150 randomly selected healthy Mongolian people from medical examination centers comprised the control group. The polymorphisms of SLCO1B1 (388A>G, 521 T > C) and ApoE (388 T > C, 526 C > t) were detected by real-time polymerase chain reaction. Combined with environmental data, the effect of gene polymorphism on coronary heart disease was explored. Results Both SLCO1B1 and ApoE polymorphisms satisfied Hardy-Weinberg equilibrium. The SLCO1B1 genotype *1a/*1b showed the highest frequency in the case group, accounting for 35.0%, while The SLCO1B1 genotype *1b/*1b showed the highest frequency in the control group, accounting for 32.0%. Allele *1b was the most commonly seen allele in both the case group and control group (57.8% and 53.7%, respectively). Meanwhile, The difference in the distribution of SLCO1B1 *1a/*15 genotype between the two groups was statistically significant (P < .05). Conclusion The results showed that the SLCO1B *1a/*15 genotype, ApoE ε3 /ε3 genotype, and ε3 allele reduced the risk of coronary disease in the Mongolian population, making them protective genes against this disease, while the ApoE ε4 allele increased the risk of coronary disease, making it a coronary disease risk factor.
Collapse
Affiliation(s)
- Rui-Bing Niu
- Department of Clinical Laboratory, Ordos Central Hospita, Ordos, Inner Mongolia, China
| | - Xiao-Xian Dong
- Department of Dermatology, Ordos Central Hospita, Ordos, Inner Mongolia, China
| | - Li-Ping Guo
- Department of Dermatology, Ordos Central Hospita, Ordos, Inner Mongolia, China
| | - Li Pan
- Department of Medicine, Ordos Central Hospita, Ordos, Inner Mongolia, China
| | - Yue-Qin Hai
- Inner Mongolia Medical University, Hohhot, China
| | - Xiao-Xiao Chen
- Baotou Medical College of Inner Mongolia, University of Science and Technology, Baotou, China
| | - Bao-Sheng Duan
- Department of Clinical Laboratory, Ordos Central Hospita, Ordos, Inner Mongolia, China
| |
Collapse
|
11
|
Kang S, Lee E, Lee H, Hwang GS, Lee J, Kim JW, Oh B, Kim JY, Kwon O. Yellow Yeast Rice Prepared Using Aspergillus terreus DSMK01 Lowers Cholesterol Levels by Stimulating Bile Salt Export Pump in Subjects with Mild-to-Moderate Hypercholesterolemia: A Randomized Controlled Trial. Mol Nutr Food Res 2021; 66:e2100704. [PMID: 34783447 DOI: 10.1002/mnfr.202100704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/01/2021] [Indexed: 01/17/2023]
Abstract
SCOPE Aspergillus terreus is an industrial microorganism used in the brewing and sauce industries. It produces monacolin K, a natural statin. The study conducted an 8-week randomized controlled trial with hypercholesterolemic subjects to examine the hypocholesterolemic effects and mechanisms of supplementation with yellow yeast rice (YYR) prepared by growing Aspergillus fungi on steamed rice. METHODS AND RESULTS YYR supplementation markedly reduced total cholesterol, LDL, and apolipoprotein B100 levels in plasma compared with the placebo. In addition, YYR induced a significantly increased ATP binding cassette subfamily B member 11 (ABCB11) gene expression compared with the placebo, indicating the role of YYR in lowering intrahepatic cholesterol availability by stimulating the bile salt export pump. Upregulation of LDL receptor (LDLR) and 3-methylglutaryl-CoA reductase (HMGCR) gene expressions provided additional evidence to support the role of YYR in reducing hepatic cholesterol availability. Plasma metabolomic profiling revealed the possibility of diminishing bile acid absorption. Finally, Spearman rank analysis showed correlations of plasma cholesterol profiles with HMGCR and LDLR gene expressions (negative) and plasma bile acids (positive). Plasma bile acids also correlated with ABCB11 (negative) and LDLR (positive) gene expressions. CONCLUSION These findings suggest that daily YYR supplementation exerted hypocholesterolemic effects in mild-to-moderate hypercholesterolemic subjects by reducing intrahepatic cholesterol availability through stimulating bile salt export pumps and inhibiting cholesterol biosynthesis.
Collapse
Affiliation(s)
- Seunghee Kang
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Eunok Lee
- Department of Nutritional Science and Food Management, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Heeyeon Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Republic of Korea
| | - Jaekyung Lee
- Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Ji Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Bumjo Oh
- Department of Family Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Ji Yeon Kim
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Oran Kwon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, 03760, Republic of Korea.,Department of Nutritional Science and Food Management, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, 03760, Republic of Korea
| |
Collapse
|
12
|
An association of ABCG8: rs11887534 polymorphism and HDL-cholesterol response to statin treatment in the Polish population. Pharmacol Rep 2021; 73:1781-1786. [PMID: 34173968 PMCID: PMC8599229 DOI: 10.1007/s43440-021-00302-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/07/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022]
Abstract
Background Variation in lipid changes in response to statin treatment is associated with genetic polymorphism. Sterolin-1, encoded by ABCG5, and sterolin-2, encoded by ABCG8, together form a sterol transporter. There are some reports indicating association of rs11887534 (ABCG8:c.55G > C) polymorphism with lipid concentrations, both prior to and after statin treatment. The aim of this study was to analyze both baseline plasma lipids and their concentrations in response to statin treatment with regard to ABCG8: rs11887534 polymorphism in Caucasian patients of Polish origin. Methods The study group consisted of 170 consecutive adult out-patients treated with atorvastatin or simvastatin for a minimum of 2 months. Concentrations of triglycerides (TG), total cholesterol (TC), LDL-cholesterol (LDL-C) and HDL-cholesterol (HDL-C) were measured before and after statin treatment. The ABCG8 polymorphism was identified by mini-sequencing genomic DNA extracted from peripheral blood leukocytes. Results There were no significant differences in regard to ABCG8 variants for baseline TG, TC, LDL-C and HDL-C as well as for TG, TC or LDL-C concentrations after statin treatment. However, patients carrying at least one C allele showed a decrease in post-statin HDL-C concentrations and the absolute and relative changes between post- and pre-statin HDL-C concentrations were negative in contrast to positive values in wild-type homozygotes. Conclusions Our results suggest that the c.55C allele of the ABCG8: rs11887534 polymorphism might be associated with decrease in HDL-cholesterol in response to statin treatment in Polish patients.
Collapse
|
13
|
Genetic variants associated with methotrexate-induced mucositis in cancer treatment: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2021; 161:103312. [PMID: 33794308 DOI: 10.1016/j.critrevonc.2021.103312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 03/08/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Methotrexate (MTX), an important chemotherapeutic agent, is often accompanied with mucositis. The occurrence and severity are unpredictable and show large interindividual variability. In this study, we review and meta-analyze previously studied genetic variants in relation to MTX-induced mucositis. We conducted a systematic search in Medline and Embase. We included genetic association studies of MTX-induced mucositis in cancer patients. A meta-analysis was conducted for single nucleotide polymorphisms (SNPs) for which at least two studies found a statistically significant association. A total of 34 SNPs were associated with mucositis in at least one study of the 57 included studies. Two of the seven SNPs included in our meta-analysis were statistically significantly associated with mucositis: MTHFR c.677C > T (recessive, grade ≥3 vs grade 0-2, OR 2.53, 95 %CI [1.48-4.32], False Discovery Rate[FDR]-corrected p-value 0.011) and MTRR c.66A > G (overdominant, grade ≥1 vs grade 0, OR 2.08, 95 %CI [1.16-3.73], FDR-corrected p-value 0.042).
Collapse
|
14
|
Ruiz-Iruela C, Candás-Estébanez B, Pintó-Sala X, Baena-Díez N, Caixàs-Pedragós A, Güell-Miró R, Navarro-Badal R, Calmarza P, Puzo-Foncilla JL, Alía-Ramos P, Padró-Miquel A. Genetic contribution to lipid target achievement with statin therapy: a prospective study. THE PHARMACOGENOMICS JOURNAL 2019; 20:494-504. [DOI: 10.1038/s41397-019-0136-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023]
|
15
|
Ali A, Levantini E, Fhu CW, Teo JT, Clohessy JG, Goggi JL, Wu CS, Chen L, Chin TM, Tenen DG. CAV1 - GLUT3 signaling is important for cellular energy and can be targeted by Atorvastatin in Non-Small Cell Lung Cancer. Am J Cancer Res 2019; 9:6157-6174. [PMID: 31534543 PMCID: PMC6735519 DOI: 10.7150/thno.35805] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/28/2019] [Indexed: 02/06/2023] Open
Abstract
Background: The development of molecular targeted therapies, such as EGFR-TKIs, has positively impacted the management of EGFR mutated NSCLC. However, patients with innate and acquired resistance to EGFR-TKIs still face limited effective therapeutic options. Statins are the most frequently prescribed anti-cholesterol agents and have been reported to inhibit the progression of various malignancies, including in lung. However, the mechanism by which statin exerts its anti-cancer effects is unclear. This study is designed to investigate the anti-proliferative effects and identify the mechanism-of-action of statins in NSCLC. Methods: In this study, the anti-tumoral properties of Atorvastatin were investigated in NSCLC utilizing cell culture system and in vivo models. Results: We demonstrate a link between elevated cellular cholesterol and TKI-resistance in NSCLC, which is independent of EGFR mutation status. Atorvastatin suppresses growth by inhibiting Cav1 expression in tumors in cell culture system and in in vivo models. Subsequent interrogations demonstrate an oncogenic physical interaction between Cav1 and GLUT3, and glucose uptake found distinctly in TKI-resistant NSCLC and this may be due to changes in the physical properties of Cav1 favoring GLUT3 binding in which significantly stronger Cav1 and GLUT3 physical interactions were observed in TKI-resistant than in TKI-sensitive NSCLC cells. Further, the differential effects of atorvastatin observed between EGFR-TKI resistant and sensitive cells suggest that EGFR mutation status may influence its actions. Conclusions: This study reveals the inhibition of oncogenic role of Cav1 in GLUT3-mediated glucose uptake by statins and highlights its potential impact to overcome NSCLC with EGFR-TKI resistance.
Collapse
|
16
|
Sun B, Li L, Zhou X. Comparative analysis of the gut microbiota in distinct statin response patients in East China. J Microbiol 2018; 56:886-892. [PMID: 30484158 DOI: 10.1007/s12275-018-8152-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/16/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Statin response shows great interindividual variations. Recently, emerging studies have shown that gut microbiota is linked to therapeutic responses to drugs, including statins. However, the association between the gut bacteria composition and statin response is still unclear. In this study, gut microbiota of 202 hyperlipidemic patients with statin sensitive (SS) response and statin resistant (SR) response in East China were investigated by high throughput sequencing to compare the gut bacteria composition and biodiversity in distinct statin response patients. Higher biodiversity was detected in Group SS than Group SR. Specifically, group SS showed significantly increased proportion of genera Lactobacillus (P = 0.001), Eubacterium (P = 0.004), Faecalibacterium (P = 0.005), and Bifidobacterium (P = 0.002) and decreased proportion of genus Clostridium (P = 0.001) compared to Group SR. The results indicated that higher gut biodiversity was associated with statin sensitive response. The increased genera Lactobacillus, Eubacterium, Faecalibacterium, Bifidobacterium, and decreased genus Clostridium in patient gut microbiota may predict patient's statin response, and hence may guide statin dosage adjustments.
Collapse
Affiliation(s)
- Baoqing Sun
- Weihai Municipal Hospital, Weihai, 264200, P. R. China
| | - Luming Li
- Weihai Municipal Hospital, Weihai, 264200, P. R. China
| | - Xinfu Zhou
- Weihai Municipal Hospital, Weihai, 264200, P. R. China.
| |
Collapse
|
17
|
Hirata RDC, Cerda A, Genvigir FDV, Hirata MH. Pharmacogenetic implications in the management of metabolic diseases in Brazilian populations. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000001005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
18
|
Slob EMA, Vijverberg SJH, Palmer CNA, Zazuli Z, Farzan N, Oliveri NMB, Pijnenburg MW, Koppelman GH, Maitland-van der Zee AH. Pharmacogenetics of inhaled long-acting beta2-agonists in asthma: A systematic review. Pediatr Allergy Immunol 2018; 29:705-714. [PMID: 29992699 DOI: 10.1111/pai.12956] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Long-acting beta2-agonists (LABA) are recommended in asthma therapy; however, not all asthma patients respond well to LABA. We performed a systematic review on genetic variants associated with LABA response in patients with asthma. METHODS Articles published until April 2017 were searched by two authors using PubMed and EMBASE. Pharmacogenetic studies in patients with asthma and LABA response as an outcome were included. RESULTS In total, 33 studies were included in this systematic review; eight focused on children (n = 6051). Nineteen studies were clinical trials, while 14 were observational studies. Studies used different outcomes to define LABA response, for example, lung function measurements (FEV1 , PEF, MMEF, FVC), exacerbations, quality of life, and asthma symptoms. Most studies (n = 30) focused on the ADRB2 gene, encoding the beta2-adrenergic receptor. Thirty studies (n = 14 874) addressed ADRB2 rs1042713, 7 ADRB2 rs1042714 (n = 1629), and 3 ADRB2 rs1800888 (n = 1892). The association of ADRB2 rs1042713 and rs1800888 with LABA response heterogeneity was successfully replicated. Other variants were only studied in three studies but not replicated. One study focused on the ADCY9 gene. Five studies and a meta-analysis found an increased risk of exacerbations in pediatrics using LABA carrying one or two A alleles (OR 1.52 [1.17; 1.99]). These results were not confirmed in adults. CONCLUSIONS ADRB2 rs1042713 variant is most consistently associated with response to LABA in children but not adults. To assess the clinical value of ADRB2 rs1042713 in children with asthma using LABA, a randomized clinical trial with well-defined outcomes is needed.
Collapse
Affiliation(s)
- Elise M A Slob
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne J H Vijverberg
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Colin N A Palmer
- Population Pharmacogenetics Group, Biomedical Research Centre, University of Dundee, Dundee, UK
| | - Zulfan Zazuli
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - Niloufar Farzan
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nadia M B Oliveri
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Mariëlle W Pijnenburg
- Department of Paediatrics, Paediatric Pulmonology & Allergology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gerard H Koppelman
- Department of Paediatric, Pulmonology & Paediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma & COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Zazuli Z, Vijverberg S, Slob E, Liu G, Carleton B, Veltman J, Baas P, Masereeuw R, Maitland-van der Zee AH. Genetic Variations and Cisplatin Nephrotoxicity: A Systematic Review. Front Pharmacol 2018; 9:1111. [PMID: 30319427 PMCID: PMC6171472 DOI: 10.3389/fphar.2018.01111] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/10/2018] [Indexed: 12/29/2022] Open
Abstract
Background: Nephrotoxicity is a notable adverse effect in cisplatin treated patients characterized by tubular injury and/or increased serum creatinine (SCr) with incidence varying from 20 to 70%. Pharmacogenomics has been shown to identify strongly predictive genetic markers to help determine which patients are more likely to experience, for example, a serious adverse drug reaction or receive optimal benefit through enhanced efficacy. Genetic variations have been reported to influence the risk of cisplatin nephrotoxicity; however, a comprehensive overview is lacking. Methods: A systematic review was performed using Pubmed, Embase and Web of Science on clinical studies that used cisplatin-based chemotherapy as treatment, had available genotyping data, and evaluated nephrotoxicity as an outcome. The quality of reporting was assessed using the STrengthening the REporting of Genetic Association Studies (STREGA) checklist. Results: Twenty-eight eligible studies were included; all were candidate gene studies. Over 300 SNPs across 135 genes were studied; 29 SNPs in 14 genes were significantly associated with cisplatin-induced nephrotoxicity. A variation in SLC22A2 rs316019, a gene involved in platinum uptake by the kidney, was associated with different measures of nephrotoxicity in four independent studies. Further, variants of ERCC1 (rs11615 and rs3212986) and ERCC2 (rs13181), two genes involved in DNA repair, were found to be positively associated with increased risks of nephrotoxicity in two independent studies. Conclusion: Three genes consistently associated with cisplatin-induced nephrotoxicity. Further research is needed to assess the biological mechanism and the clinical value of modifying treatment based on SLCC22A2 and ERCC1/2 genotypes.
Collapse
Affiliation(s)
- Zulfan Zazuli
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - Susanne Vijverberg
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Elise Slob
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Hospital-University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, Toronto, ON, Canada
| | - Bruce Carleton
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Joris Veltman
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Paul Baas
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, Netherlands
| | | |
Collapse
|
20
|
Rocha KCE, Pereira BMV, Rodrigues AC. An update on efflux and uptake transporters as determinants of statin response. Expert Opin Drug Metab Toxicol 2018; 14:613-624. [PMID: 29842801 DOI: 10.1080/17425255.2018.1482276] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Statins are used in the treatment of dyslipidemia promoting primary and secondary prevention against detrimental cardiovascular events. ATP-binding cassette (ABC) and solute carrier (SLC) membrane transporters transport statins across the cell membrane. Differences in drug transporter tissue expression and activity contribute to variability in statin pharmacokinetics (PK) and response. Areas covered: The purpose of this review is to discuss factors impacting transporter expression and the effect this has on statin efficacy and safety. Previous studies have demonstrated that genetic polymorphisms, drug-drug interactions (DDI), nuclear receptors, and microRNAs affect statin PK and pharmacodynamics. Expert opinion: Genetic variants of ABCG2 and SLCO1B1 transporters affect statin PK and, as a result, the intended lipid-lowering response. However, the effect size is small, limiting its applicability in clinical practice. Furthermore, genetic variants do not totally explain the observed intervariability in statin response. Thus, it is likely that transcriptional and post-transcriptional regulation of drug transporters are also highly involved. Further studies are required to understand the contribution of each of these new factors in statin disposition and toxicity.
Collapse
Affiliation(s)
- Karina Cunha E Rocha
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , SP , Brazil
| | - Beatriz Maria Veloso Pereira
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , SP , Brazil
| | - Alice Cristina Rodrigues
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , SP , Brazil
| |
Collapse
|
21
|
Taylor BA, Panza G, Ballard KD, White CM, Thompson PD. Creatine supplementation does not alter the creatine kinase response to eccentric exercise in healthy adults on atorvastatin. J Clin Lipidol 2018; 12:1305-1312. [PMID: 29945780 DOI: 10.1016/j.jacl.2018.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Serum creatine kinase (CK) levels are higher after eccentric, muscle-damaging exercise in statin-treated patients. This could contribute to the increased statin-associated muscle symptoms reported in physically active individuals. OBJECTIVE We tested the hypothesis in this pilot study that creatine (Cr) monohydrate supplementation would reduce the CK response to eccentric exercise in patients using statins to determine if Cr supplementation could be a strategy to mitigate statin-associated muscle symptoms in physically active individuals. METHODS Healthy, nonsmoking men (n = 5) and women (n = 14) were randomized to Cr monohydrate = atorvastatin 80 mg + 10 g Cr monohydrate (n = 10, age = 60 ± 7 years) or to placebo (PL) = atorvastatin 80 mg + PL (n = 9, age = 52 ± 6 years). After 4 weeks of treatment, subjects performed 45 minutes of eccentric exercise (downhill walking at a -15% grade). Serum CK levels, muscle soreness (visual analog scale after two squats), and muscle pain severity and interference (using the brief pain inventory) were measured before and after 4 weeks of treatment, and then for 4 consecutive days after downhill walking. Vitamin D, or serum 25(OH)D, was also measured at baseline. RESULTS The PL group was younger (P = .01) but not otherwise different in blood lipids, vitamin D, CK, muscle visual analog scale, and pain scores before (all P > .21) or after (all P > .12) treatment. CK increased in all subjects after downhill walking (P < .01), but neither the relative peak change (expressed as group mean difference with 95% confidence intervals: 43.52% [-196.41, 283.45]) nor the absolute peak change (67.38 U/L [-121.55, 256.31]) relative to baseline was different between groups (P = .46 and .71, respectively). A similar lack of treatment effect was observed for muscle soreness (11.03 mm [-9.49, 31.55]), pain severity (0.77 pts [-0.95, 2.50]), and pain interference (1.02 pts [-1.25, 3.29]) with P-values for group comparisons = 0.27, 0.36, and 0.35, respectively. However, subjects with "insufficient" Vitamin D < 30 ng/mL (n = 10) had an ∼2-fold greater CK increase with eccentric exercise (nominal P-value = .04) than subjects with higher vitamin D levels. CONCLUSION Cr monohydrate did not reduce CK increases after exercise in statin-treated subjects. We did observe that low vitamin D levels are associated with a greater CK response to eccentric exercise in statin-treated subjects.
Collapse
Affiliation(s)
- Beth A Taylor
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA; Department of Kinesiology, University of Connecticut, Storrs, CT, USA.
| | - Gregory Panza
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA; Department of Kinesiology, University of Connecticut, Storrs, CT, USA
| | - Kevin D Ballard
- Department of Kinesiology and Health, Miami University, Oxford, OH, USA
| | - C Michael White
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA; University of Connecticut School of Medicine, Farmington, CT, USA
| | - Paul D Thompson
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA; University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
22
|
|
23
|
Abstract
Blood lipids are important modifiable risk factors for coronary heart disease and various drugs have been developed to target lipid fractions. Considerable efforts have been made to identify genetic variants that modulate responses to drugs in the hope of optimizing their use. Pharmacogenomics and new biotechnologies now allow for meaningful integration of human genetic findings and therapeutic development for increased efficiency and precision of lipid-lowering drugs. Polygenic predictors of disease risk are also changing how patient populations can be stratified, enabling targeted therapeutic interventions to patients more likely to derive the highest benefit, marking a shift from single variant to genomic approaches in pharmacogenomics.
Collapse
Affiliation(s)
- Marc-André Legault
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada.,Université de Montréal, Faculté de médecine, Montreal, QC, H3T 1J4, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, H1T 1C8, QC, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada.,Université de Montréal, Faculté de médecine, Montreal, QC, H3T 1J4, Canada
| | - Marie-Pierre Dubé
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada.,Université de Montréal, Faculté de médecine, Montreal, QC, H3T 1J4, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, H1T 1C8, QC, Canada
| |
Collapse
|
24
|
Soko ND, Masimirembwa C, Dandara C. Pharmacogenomics of Rosuvastatin: A Glocal (Global+Local) African Perspective and Expert Review on a Statin Drug. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 20:498-509. [PMID: 27631189 DOI: 10.1089/omi.2016.0114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The incidence of cardiovascular diseases (CVDs) in African populations residing in the African continent is on the rise fueled by both a steady increase in CVD risk factors and comorbidities such as human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS), tuberculosis, and parasitic diseases such as bilharzia. Statins are recommended together with lifestyle changes in the treatment of hypercholesterolemia and overall reduction of cardiovascular events. Rosuvastatin in particular is an attractive candidate in the management of CVDs in African populations often plagued with multimorbidities owing to both its potency and low drug-to-drug interaction potential. In this expert review, we describe the pharmacogenetics of rosuvastatin and how it may instrumentally affect the African populations. We describe polymorphisms in the candidate genes, ABCG2, SLCO1B1, CYP2C9, APOE, PCSK9, LDLR, LPA, and HMGCR, and their role in the potency and safety of rosuvastatin therapy. We report on qualitative and quantitative differences in the distribution of genetic variants that affect efficacy and toxicity of rosuvastatin. These differences are observed across world populations (Caucasian, European, and Asian) as well as within African populations. Finally, we advocate for extensive pharmacogenetic studies in African populations that take into account the genetic diversity of intra-African ethnic groups and the genetic differences between African populations and other global populations, with a collaborative and collective aim to provide effective and safe use of rosuvastatin in management of CVD in Africa. Our key thesis presented in this innovation field analysis is that rosuvastatin precision medicine can serve as a veritable Glocal (Global and Local) model to offer pharmacogenetic-guided optimal therapeutics for the public in both developing and developed regions of the world.
Collapse
Affiliation(s)
- Nyarai D Soko
- 1 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, and University of Cape Town , Cape Town, South Africa
| | - Collen Masimirembwa
- 2 African Institute of Biomedical Science and Technology (AiBST) , Wilkins Hospital, Harare, Zimbabwe .,3 Clinical Pharmacology, Department of Medicine, University of Cape Town , Cape Town, South Africa
| | - Collet Dandara
- 1 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, and University of Cape Town , Cape Town, South Africa
| |
Collapse
|
25
|
Polymorphisms in CYP450 Genes and the Therapeutic Effect of Atorvastatin on Ischemic Stroke: A Retrospective Cohort Study in Chinese Population. Clin Ther 2018; 40:469-477.e2. [DOI: 10.1016/j.clinthera.2018.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/13/2018] [Accepted: 02/03/2018] [Indexed: 12/19/2022]
|
26
|
Smit RAJ, Noordam R, le Cessie S, Trompet S, Jukema JW. A critical appraisal of pharmacogenetic inference. Clin Genet 2018; 93:498-507. [PMID: 29136278 DOI: 10.1111/cge.13178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/25/2017] [Accepted: 11/09/2017] [Indexed: 01/06/2023]
Abstract
In essence, pharmacogenetic research is aimed at discovering variants of importance to gene-treatment interaction. However, epidemiological studies are rarely set up with this goal in mind. It is therefore of great importance that researchers clearly communicate which assumptions they have had to make, and which inherent limitations apply to the interpretation of their results. This review discusses considerations of, and the underlying assumptions for, utilizing different response phenotypes and study designs popular in pharmacogenetic research to infer gene-treatment interaction effects, with a special focus on those dealing with of clinical effects of drug treatment.
Collapse
Affiliation(s)
- R A J Smit
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands.,Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - R Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - S le Cessie
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - S Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands.,Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
27
|
Rafeeq MM, Habib HS, Murad HAS, Gari MA, Gazzaz ZJ. Effect of genetic polymorphisms in SREBF-SCAP pathway on therapeutic response to rosuvastatin in Saudi metabolic syndrome patients. Pharmacogenomics 2018; 19:185-196. [PMID: 29318930 DOI: 10.2217/pgs-2017-0181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Genetic variants contribute to statins' therapeutic variability. SREBF-SCAP pathway is a key player in lipid homeostasis. Hence, effect of SREBF-SCAP polymorphisms on therapeutic response was studied. PATIENTS & METHODS Metabolic syndrome patients of either sex were prescribed rosuvastatin 10 mg for 24 weeks. Clinical, anthropometric and lipid measurements were done before and after treatment. Genotyping was done by pyrosequencing. RESULTS & CONCLUSION No associations of SCAP and SREBF-1a genotypes with baseline lipids but significant associations with lipid reductions were observed. Significant effect of SCAP (GG; B = -8.16, p = 0.001); SREBF-1a (GG; B = -7.47, p = 0.001) and SREBF-1a (-delG; B = -7.42, p = 0.001) was observed on total cholesterol reduction. Additive trend was found between SCAP genotypes and lipid reductions. A total of 88% responders have SCAP 'G' allele (p = 0.001). Patients carrying SCAP (GG) and SREBF-1a (GG and -delG) have 9.5-, 8.6- and 14.6-times more likelihood of being responders (p < 0.05). 'G' allele in SCAP and SREBF-1a is significant predictor of rosuvastatin response.
Collapse
Affiliation(s)
- Misbahuddin Mohd Rafeeq
- Department of Pharmacology, Faculty of Medicine, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hamed Said Habib
- Department of Paediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hussam Aly Sayed Murad
- Department of Pharmacology, Faculty of Medicine, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia & Faculty of Medicine, Ainshams University, Cairo, Egypt
| | - Mamdouh Abdullah Gari
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zohair Jamil Gazzaz
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
28
|
Systems Pharmacology Dissection of Cholesterol Regulation Reveals Determinants of Large Pharmacodynamic Variability between Cell Lines. Cell Syst 2017; 5:604-619.e7. [PMID: 29226804 PMCID: PMC5747350 DOI: 10.1016/j.cels.2017.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 08/17/2017] [Accepted: 11/02/2017] [Indexed: 01/06/2023]
Abstract
In individuals, heterogeneous drug-response phenotypes result from a complex interplay of dose, drug specificity, genetic background, and environmental factors, thus challenging our understanding of the underlying processes and optimal use of drugs in the clinical setting. Here, we use mass-spectrometry-based quantification of molecular response phenotypes and logic modeling to explain drug-response differences in a panel of cell lines. We apply this approach to cellular cholesterol regulation, a biological process with high clinical relevance. From the quantified molecular phenotypes elicited by various targeted pharmacologic or genetic treatments, we generated cell-line-specific models that quantified the processes beneath the idiotypic intracellular drug responses. The models revealed that, in addition to drug uptake and metabolism, further cellular processes displayed significant pharmacodynamic response variability between the cell lines, resulting in cell-line-specific drug-response phenotypes. This study demonstrates the importance of integrating different types of quantitative systems-level molecular measurements with modeling to understand the effect of pharmacological perturbations on complex biological processes.
Collapse
|
29
|
Jansen ME, Rigter T, Rodenburg W, Fleur TMC, Houwink EJF, Weda M, Cornel MC. Review of the Reported Measures of Clinical Validity and Clinical Utility as Arguments for the Implementation of Pharmacogenetic Testing: A Case Study of Statin-Induced Muscle Toxicity. Front Pharmacol 2017; 8:555. [PMID: 28878673 PMCID: PMC5572384 DOI: 10.3389/fphar.2017.00555] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/07/2017] [Indexed: 12/19/2022] Open
Abstract
Advances from pharmacogenetics (PGx) have not been implemented into health care to the expected extent. One gap that will be addressed in this study is a lack of reporting on clinical validity and clinical utility of PGx-tests. A systematic review of current reporting in scientific literature was conducted on publications addressing PGx in the context of statins and muscle toxicity. Eighty-nine publications were included and information was selected on reported measures of effect, arguments, and accompanying conclusions. Most authors report associations to quantify the relationship between a genetic variation an outcome, such as adverse drug responses. Conclusions on the implementation of a PGx-test are generally based on these associations, without explicit mention of other measures relevant to evaluate the test's clinical validity and clinical utility. To gain insight in the clinical impact and select useful tests, additional outcomes are needed to estimate the clinical validity and utility, such as cost-effectiveness.
Collapse
Affiliation(s)
- Marleen E Jansen
- Section Community Genetics, Department of Clinical Genetics and Amsterdam Public Health Research Institute, VU University Medical CenterAmsterdam, Netherlands.,Centre for Health Protection, National Institute for Public Health and the EnvironmentBilthoven, Netherlands
| | - T Rigter
- Section Community Genetics, Department of Clinical Genetics and Amsterdam Public Health Research Institute, VU University Medical CenterAmsterdam, Netherlands.,Centre for Health Protection, National Institute for Public Health and the EnvironmentBilthoven, Netherlands
| | - W Rodenburg
- Centre for Health Protection, National Institute for Public Health and the EnvironmentBilthoven, Netherlands
| | - T M C Fleur
- Section Community Genetics, Department of Clinical Genetics and Amsterdam Public Health Research Institute, VU University Medical CenterAmsterdam, Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht UniversityUtrecht, Netherlands
| | - E J F Houwink
- Section Community Genetics, Department of Clinical Genetics and Amsterdam Public Health Research Institute, VU University Medical CenterAmsterdam, Netherlands
| | - M Weda
- Centre for Health Protection, National Institute for Public Health and the EnvironmentBilthoven, Netherlands
| | - Martina C Cornel
- Section Community Genetics, Department of Clinical Genetics and Amsterdam Public Health Research Institute, VU University Medical CenterAmsterdam, Netherlands
| |
Collapse
|
30
|
Mehdar A, Hegele RA, Kim RB, Gryn SE. Statin therapy: time for a precision medicine approach? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1356685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Albayda Mehdar
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Robert A Hegele
- Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON, Canada
| | - Richard B. Kim
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Steven E. Gryn
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
31
|
Abstract
Pharmacogenomics (PGx), a substantial component of "personalized medicine", seeks to understand each individual's genetic composition to optimize drug therapy -- maximizing beneficial drug response, while minimizing adverse drug reactions (ADRs). Drug responses are highly variable because innumerable factors contribute to ultimate phenotypic outcomes. Recent genome-wide PGx studies have provided some insight into genetic basis of variability in drug response. These can be grouped into three categories. [a] Monogenic (Mendelian) traits include early examples mostly of inherited disorders, and some severe (idiosyncratic) ADRs typically influenced by single rare coding variants. [b] Predominantly oligogenic traits represent variation largely influenced by a small number of major pharmacokinetic or pharmacodynamic genes. [c] Complex PGx traits resemble most multifactorial quantitative traits -- influenced by numerous small-effect variants, together with epigenetic effects and environmental factors. Prediction of monogenic drug responses is relatively simple, involving detection of underlying mutations; due to rarity of these events and incomplete penetrance, however, prospective tests based on genotype will have high false-positive rates, plus pharmacoeconomics will require justification. Prediction of predominantly oligogenic traits is slowly improving. Although a substantial fraction of variation can be explained by limited numbers of large-effect genetic variants, uncertainty in successful predictions and overall cost-benefit ratios will make such tests elusive for everyday clinical use. Prediction of complex PGx traits is almost impossible in the foreseeable future. Genome-wide association studies of large cohorts will continue to discover relevant genetic variants; however, these small-effect variants, combined, explain only a small fraction of phenotypic variance -- thus having limited predictive power and clinical utility.
Collapse
Affiliation(s)
- Ge Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, United States.
| | - Daniel W Nebert
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, United States; Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati School of Medicine, Cincinnati, OH 45267-0056, United States.
| |
Collapse
|
32
|
|
33
|
Rumyantsev NA, Kukes VG, Kazakov RE, Rumyantsev AA, Sychev DA. [Use of pharmacogenetic testing to prevent adverse drug reactions during statin therapy]. TERAPEVT ARKH 2017; 89:82-87. [PMID: 28252633 DOI: 10.17116/terarkh201789182-87] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The number of patients receiving statins increases every year and due to the fact that they should take statins during their lives, the problem of their safety use comes to the forefront. The paper analyzes the safety of using the medications of this group and discusses the diagnosis of myopathies induced by statins and the occurrence of immune-mediated statin myopathies. It considers a personalized approach to prescribing statins, analyzes Russian and foreign experience in using pharmacogenetics to reduce the risk of myopathies, publishes the results of the authors' experience in clinically introducing pharmacogenetic testing at hospitals, and analyzes the long-term results of determining the polymorphism of the SLCO1B1 gene for the prediction of the risk of adverse events when using statins and estimating patient compliance to prescribed treatment.
Collapse
Affiliation(s)
- N A Rumyantsev
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia; Research Center for Examination of Medical Products, Ministry of Health of Russia, Moscow, Russia
| | - V G Kukes
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia; Research Center for Examination of Medical Products, Ministry of Health of Russia, Moscow, Russia
| | - R E Kazakov
- Research Center for Examination of Medical Products, Ministry of Health of Russia, Moscow, Russia
| | - A A Rumyantsev
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - D A Sychev
- Russian Medical Academy of Postgraduate Education, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
34
|
Abstract
Statins are used widely in primary and secondary prevention of cardiovascular disease; a treatment effect that has long been thought to be due to their cholesterol-lowering properties. However, statins also have a wide range of anti-inflammatory effects independent of their lipid-lowering mechanisms. In depression, low-grade inflammation is a replicated finding, and several studies have shown antidepressant properties of diverse anti-inflammatory drugs. Large observational studies have suggested reduced risks of depression amongst those taking statins, an effect that is thought to be explained by the anti-inflammatory properties of this class of drugs. Also, preliminary randomized controlled trials (RCTs) have indicated that statins may have adjunctive antidepressant effects when used as add-on treatment to selective serotonin reuptake inhibitors (SSRIs). However, the RCTs were small and limited by low generalizability, and some early observational studies have pointed towards potential neuropsychiatric adverse effects of statin treatment. Nevertheless, based on the good tolerability and general safety of the statins, researchers are currently investigating the potential antidepressant properties of these agents. The present review aims to give an overview on the potential antidepressant effects of statins based on their anti-inflammatory properties, covering topics such as safety versus treatment effects, potential mechanisms of action and the possibility of targeted treatment (precision medicine).
Collapse
Affiliation(s)
- Ole Köhler-Forsberg
- Psychosis Research Unit, Aarhus University Hospital, Skovagervej 2, 8240, Risskov, Denmark. .,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark. .,Mental Health Centre Copenhagen, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Christiane Gasse
- National Centre for Register-Based Research (NCRR), Aarhus University, Aarhus, Denmark.,iPSYCH, The Lundbeck Initiative for Integrated Research in Psychiatry, Aarhus, Denmark
| | - Michael Berk
- Deakin University, School of Medicine, IMPACT Strategic Research Centre, Geelong, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia.,The Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia.,Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia
| | - Søren Dinesen Østergaard
- Psychosis Research Unit, Aarhus University Hospital, Skovagervej 2, 8240, Risskov, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
35
|
de With SAJ, Pulit SL, Staal WG, Kahn RS, Ophoff RA. More than 25 years of genetic studies of clozapine-induced agranulocytosis. THE PHARMACOGENOMICS JOURNAL 2017; 17:304-311. [PMID: 28418011 DOI: 10.1038/tpj.2017.6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/23/2016] [Accepted: 01/18/2017] [Indexed: 12/18/2022]
Abstract
Clozapine is one of the most effective atypical antipsychotic drugs prescribed to patients with treatment-resistant schizophrenia. Approximately 1% of patients experience potential life-threatening adverse effects in the form of agranulocytosis, greatly hindering its applicability in clinical practice. The etiology of clozapine-induced agranulocytosis (CIA) remains unclear, but is thought to be a heritable trait. We reviewed the genetic studies of CIA published thus far. One recurrent finding from early candidate gene study to more recent genome-wide analysis is that of the involvement of human leukocyte antigen locus. We conclude that CIA is most likely a complex, polygenic trait, which may hamper efforts to the development of a genetic predictor test with clinical relevance. To decipher the genetic architecture of CIA, it is necessary to apply more rigorous standards of phenotyping and study much larger sample sizes.
Collapse
Affiliation(s)
- S A J de With
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S L Pulit
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - W G Staal
- Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands.,Department of Psychiatry, Radboud University Nijmegen Medical Center and Karakter, Center for Child and Adolescent Psychiatry, Nijmegen, The Netherlands
| | - R S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R A Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,UCLA Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| |
Collapse
|
36
|
Arrigoni E, Del Re M, Fidilio L, Fogli S, Danesi R, Di Paolo A. Pharmacogenetic Foundations of Therapeutic Efficacy and Adverse Events of Statins. Int J Mol Sci 2017; 18:ijms18010104. [PMID: 28067828 PMCID: PMC5297738 DOI: 10.3390/ijms18010104] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 12/11/2022] Open
Abstract
Background: In the era of precision medicine, more attention is paid to the search for predictive markers of treatment efficacy and tolerability. Statins are one of the classes of drugs that could benefit from this approach because of their wide use and their incidence of adverse events. Methods: Literature from PubMed databases and bibliography from retrieved publications have been analyzed according to terms such as statins, pharmacogenetics, epigenetics, toxicity and drug–drug interaction, among others. The search was performed until 1 October 2016 for articles published in English language. Results: Several technical and methodological approaches have been adopted, including candidate gene and next generation sequencing (NGS) analyses, the latter being more robust and reliable. Among genes identified as possible predictive factors associated with statins toxicity, cytochrome P450 isoforms, transmembrane transporters and mitochondrial enzymes are the best characterized. Finally, the solute carrier organic anion transporter family member 1B1 (SLCO1B1) transporter seems to be the best target for future studies. Moreover, drug–drug interactions need to be considered for the best approach to personalized treatment. Conclusions: Pharmacogenetics of statins includes several possible genes and their polymorphisms, but muscular toxicities seem better related to SLCO1B1 variant alleles. Their analysis in the general population of patients taking statins could improve treatment adherence and efficacy; however, the cost–efficacy ratio should be carefully evaluated.
Collapse
Affiliation(s)
- Elena Arrigoni
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Leonardo Fidilio
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Stefano Fogli
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy.
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Antonello Di Paolo
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| |
Collapse
|
37
|
Rafeeq MM, Ahmad F, Rahman SZ, Siddiqi SS, Shakil S. Effect of an SNP in SCAP gene on lipid-lowering response to rosuvastatin in Indian patients with metabolic syndrome. Pharmacogenomics 2016; 17:2015-2024. [PMID: 27885915 DOI: 10.2217/pgs.16.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
AIM Statins treat dyslipidemia associated with metabolic syndrome. Genetic factors contribute to variable response. Sterol regulatory element-binding factors cleavage-activating protein (SCAP) pathway regulates lipid homeostasis, so effect of SNP in SCAP gene on rosuvastatin response was studied. MATERIALS & METHODS Metabolic syndrome patients with low-density lipoprotein-cholesterol ≥130 mg/dl, were prescribed rosuvastatin 5 mg for 3 months. Lipids were measured initially and finally, and genotyping done. RESULTS & CONCLUSION Sixty-three patients completed the study. Twenty-three were homozygous for AA while 40 were heterozygous. Significant association was found between post-treatment lipid values and SCAP genotypes but not with baseline values. Cholesterol (p = 0.002) and low-density lipoprotein-cholesterol (p = 0.008) were significantly reduced in patients carrying G allele as compared with AA. There was a significant effect of G allele on cholesterol reduction (p = 0.043). Out of total responders (achieving >23.58% total cholesterol reduction), 80.5% were 2386G carriers (GG+GA) and only 19.5% were homozygous for A allele (p = 0.0048). SCAP 2386A>G gene polymorphism is a significant predictor of hypolipidemic response.
Collapse
Affiliation(s)
- Misbahuddin M Rafeeq
- Department of Pharmacology, Rabigh College of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Farida Ahmad
- Department of Pharmacology, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, India
| | - Syed Z Rahman
- Department of Pharmacology, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, India
| | - Sheelu S Siddiqi
- Rajiv Gandhi Centre for Diabetes & Endocrinology, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, India
| | - Shazi Shakil
- Centre for Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
38
|
Kitzmiller JP, Luzum JA, Dauki A, Krauss RM, Medina MW. Candidate-Gene Study of Functional Polymorphisms in SLCO1B1 and CYP3A4/5 and the Cholesterol-Lowering Response to Simvastatin. Clin Transl Sci 2016; 10:172-177. [PMID: 28482130 PMCID: PMC5421731 DOI: 10.1111/cts.12432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/18/2016] [Indexed: 01/23/2023] Open
Abstract
Cholesterol‐lowering response to 40 mg simvastatin daily for 6 weeks was examined for associations with common genetic polymorphisms in key genes affecting simvastatin metabolism (CYP3A4 and CYP3A5) and transport (SLCO1B1). In white people (n = 608), SLCO1B1 521C was associated with lesser reductions of total and low‐density lipoprotein cholesterol. Associations between SLCO1B1 521C and cholesterol response were not detected in African Americans (n = 333). Associations between CYP3A4*22 or CYP3A5*3 and cholesterol response were not detected in either race, and no significant race‐gene or gene‐gene interactions were detected. As several of the analyses may have been underpowered (especially the analyses in the African American cohort), the findings not suggesting an association should not be considered conclusive and warrant further investigation. The finding regarding SLCO1B1 521C in whites was consistent with several previous reports. SLCO1B1 521C resulted in a diminished cholesterol‐lowering response, but a marginal effect size limits utility for predicting simvastatin response.
Collapse
Affiliation(s)
- J P Kitzmiller
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - J A Luzum
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - A Dauki
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, Ohio, USA.,College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - R M Krauss
- Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - M W Medina
- Children's Hospital Oakland Research Institute, Oakland, California, USA
| |
Collapse
|
39
|
Kitzmiller JP, Mikulik EB, Dauki AM, Murkherjee C, Luzum JA. Pharmacogenomics of statins: understanding susceptibility to adverse effects. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2016; 9:97-106. [PMID: 27757045 PMCID: PMC5055044 DOI: 10.2147/pgpm.s86013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Statins are a cornerstone of the pharmacologic treatment and prevention of atherosclerotic cardiovascular disease. Atherosclerotic disease is a predominant cause of mortality and morbidity worldwide. Statins are among the most commonly prescribed classes of medications, and their prescribing indications and target patient populations have been significantly expanded in the official guidelines recently published by the American and European expert panels. Adverse effects of statin pharmacotherapy, however, result in significant cost and morbidity and can lead to nonadherence and discontinuation of therapy. Statin-associated muscle symptoms occur in ~10% of patients on statins and constitute the most commonly reported adverse effect associated with statin pharmacotherapy. Substantial clinical and nonclinical research effort has been dedicated to determining whether genetics can provide meaningful insight regarding an individual patient’s risk of statin adverse effects. This contemporary review of the relevant clinical research on polymorphisms in several key genes that affect statin pharmacokinetics (eg, transporters and metabolizing enzymes), statin efficacy (eg, drug targets and pathways), and end-organ toxicity (eg, myopathy pathways) highlights several promising pharmacogenomic candidates. However, SLCO1B1 521C is currently the only clinically relevant pharmacogenetic test regarding statin toxicity, and its relevance is limited to simvastatin myopathy.
Collapse
Affiliation(s)
| | - Eduard B Mikulik
- Department of Biological Chemistry and Pharmacology, College of Medicine
| | - Anees M Dauki
- College of Pharmacy, The Ohio State University, Columbus, OH
| | | | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
40
|
|
41
|
Precision Medicine, Cardiovascular Disease and Hunting Elephants. Prog Cardiovasc Dis 2016; 58:651-60. [DOI: 10.1016/j.pcad.2016.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/14/2023]
|