1
|
Palmen R, Walton M, Wagner J. Pediatric flecainide pharmacogenomics: a roadmap to delivering precision-based care to pediatrics arrhythmias. Front Pharmacol 2024; 15:1477485. [PMID: 39741635 PMCID: PMC11686437 DOI: 10.3389/fphar.2024.1477485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Flecainide acetate is a Class 1c anti-arrhythmic with a potent sodium voltage gated channel blockade which is utilized for the second-line treatment of tachyarrhythmias in children and adults. Given its narrow therapeutic index, the individualization of drug therapy is of utmost importance for clinicians. Despite efforts to improve anti-arrhythmic drug therapy, there remain knowledge gaps regarding the impact of variation in the genes relevant to flecainide's disposition and response. This variability is compounded in developing children whose drug disposition and response pathways may remain immature. The purpose of this comprehensive review is to outline flecainide's disposition and response pathways while simultaneously highlighting opportunities for prospective investigation in the pediatric population.
Collapse
Affiliation(s)
- Ronald Palmen
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Mollie Walton
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| | - Jonathan Wagner
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| |
Collapse
|
2
|
An Integrated Transcriptomic Approach to Identify Molecular Markers of Calcineurin Inhibitor Nephrotoxicity in Pediatric Kidney Transplant Recipients. Int J Mol Sci 2021; 22:ijms22115414. [PMID: 34063776 PMCID: PMC8196602 DOI: 10.3390/ijms22115414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 01/29/2023] Open
Abstract
Calcineurin inhibitors are highly efficacious immunosuppressive agents used in pediatric kidney transplantation. However, calcineurin inhibitor nephrotoxicity (CNIT) has been associated with the development of chronic renal allograft dysfunction and decreased graft survival. This study evaluated 37 formalin-fixed paraffin-embedded biopsies from pediatric kidney transplant recipients using gene expression profiling. Normal allograft samples (n = 12) served as negative controls and were compared to biopsies exhibiting CNIT (n = 11). The remaining samples served as positive controls to validate CNIT marker specificity and were characterized by other common causes of graft failure such as acute rejection (n = 7) and interstitial fibrosis/tubular atrophy (n = 7). MiRNA profiles served as the platform for data integration. Oxidative phosphorylation and mitochondrial dysfunction were the top molecular pathways associated with overexpressed genes in CNIT samples. Decreased ATP synthesis was identified as a significant biological function in CNIT, while key toxicology pathways included NRF2-mediated oxidative stress response and increased permeability transition of mitochondria. An integrative analysis demonstrated a panel of 13 significant miRNAs and their 33 CNIT-specific gene targets involved with mitochondrial activity and function. We also identified a candidate panel of miRNAs/genes, which may serve as future molecular markers for CNIT diagnosis as well as potential therapeutic targets.
Collapse
|
3
|
Giangreco NP, Elias JE, Tatonetti NP. No population left behind: Improving paediatric drug safety using informatics and systems biology. Br J Clin Pharmacol 2020; 88:1464-1470. [PMID: 33332641 PMCID: PMC8209126 DOI: 10.1111/bcp.14705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/26/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Adverse drugs effects (ADEs) in children are common and may result in disability and death. The current paediatric drug safety landscape, including clinical trials, is limited as it rarely includes children and relies on extrapolation from adults. Children are not small adults but go through an evolutionarily conserved and physiologically dynamic process of growth and maturation. Novel quantitative approaches, integrating observations from clinical trials and drug safety databases with dynamic mechanisms, can be used to systematically identify ADEs unique to childhood. In this perspective, we discuss three critical research directions using systems biology methodologies and novel informatics to improve paediatric drug safety, namely child versus adult drug safety profiles, age-dependent drug toxicities and genetic susceptibility of ADEs across childhood. We argue that a data-driven framework that leverages observational data, biomedical knowledge and systems biology modelling will reveal previously unknown mechanisms of pediatric adverse drug events and lead to improved paediatric drug safety.
Collapse
Affiliation(s)
- Nicholas P Giangreco
- Department of Biomedical Informatics and Systems Biology, Columbia University, New York, NY, USA
| | - Jonathan E Elias
- Department of Pediatrics, Instructor in Pediatrics, Assistant Medical Director of Information Services, Weill Cornell Medical & NYP Weill Cornell Medical Center, New York, NY, USA
| | - Nicholas P Tatonetti
- Department of Biomedical Informatics and Systems Biology, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Mansouri M, Yuan B, Ross CJD, Carleton BC, Ester M. HUME: large-scale detection of causal genetic factors of adverse drug reactions. Bioinformatics 2018; 34:4274-4283. [PMID: 29931042 DOI: 10.1093/bioinformatics/bty475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/14/2018] [Indexed: 11/12/2022] Open
Abstract
Motivation Adverse drug reactions are one of the major factors that affect the wellbeing of patients and financial costs of healthcare systems. Genetic variations of patients have been shown to be a key factor in the occurrence and severity of many ADRs. However, the large number of confounding drugs and genetic biomarkers for each adverse reaction case demands a method that evaluates all potential genetic causes of ADRs simultaneously. Results To address this challenge, we propose HUME, a multi-phase algorithm that recommends genetic factors for ADRs that are causally supported by the patient record data. HUME consists of the construction of a network from co-prevalence between significant genetic biomarkers and ADRs, a link score phase for predicting candidate relations based on the Adamic-Adar measure, and a causal refinement phase based on multiple hypothesis testing of quasi experimental designs for evaluating evidence and counter evidence of candidate relations in the patient records. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mehrdad Mansouri
- Department of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Bowei Yuan
- Department of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Colin J D Ross
- Child and Family Research Institute, Children's and Women's Health Research Centre of British Columbia, Vancouver, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Bruce C Carleton
- Child and Family Research Institute, Children's and Women's Health Research Centre of British Columbia, Vancouver, British Columbia, Canada.,Department of Paediatrics, Faculty of Pharmaceutical Sciences, Pharmaceutical Outcomes Programme, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Ester
- Department of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
5
|
Khan LM, Kamel FO, Alkreathy HM, Al-Harthi SE, Saadah OI, Osman AMM, Allibaih MA. Benefits of Medication Antidote Signals for the Detection of Potential Adverse Drug Reactions over Contemporary Methods of Pharmacovigilance in Hospitalized Children. INT J PHARMACOL 2016. [DOI: 10.3923/ijp.2017.64.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
6
|
Pharmacogenomic incidental findings in 308 families: The NIH Undiagnosed Diseases Program experience. Genet Med 2016; 18:1303-1307. [PMID: 27253732 PMCID: PMC5133159 DOI: 10.1038/gim.2016.47] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/08/2016] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Using SNP chip and exome sequence data from individuals participating in the NIH Undiagnosed Diseases Program (UDP), we evaluated the number and therapeutic informativeness of incidental pharmacogenetic variants. METHODS Pharmacogenomics Knowledgebase (PharmGKB) annotated sequence variants were identified in 1,101 individuals. Medication records of participants were used to identify individuals prescribed medications for which a genetic variant might alter efficacy. RESULTS 395 sequence variants, including 19 PharmGKB 1A and 1B variants, were identified in SNP chip sequence data and 388 variants, including 21 PharmGKB 1A and 1B variants, were identified in the exome sequence data. Nine participants had incidental pharmacogenetic variants associated with altered efficacy of a prescribed medication. CONCLUSIONS Despite the small size of the NIH UDP patient cohort, we identified pharmacogenetic incidental findings potentially useful for guiding therapy. Consequently, groups conducting clinical genomic studies might consider reporting of pharmacogenetic incidental findings.
Collapse
|
7
|
P450 oxidoreductase *28 (POR*28) and tacrolimus disposition in pediatric kidney transplant recipients--a pilot study. Ther Drug Monit 2015; 36:152-8. [PMID: 24089076 DOI: 10.1097/ftd.0b013e3182a3f282] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Both age and CYP3A5 genotype are important determinants of tacrolimus disposition in pediatric kidney transplant recipients. In a recent study in adults, POR*28 was associated with increased dosing requirements early after transplant of CYP3A5-expressing kidney transplant recipients. The authors aimed to evaluate the additional contribution of POR*28 to early tacrolimus disposition in pediatric kidney transplant recipients. METHODS Retrospective data of 43 pediatric kidney transplant recipients up to 14 days posttransplant were evaluated on tacrolimus dose and tacrolimus predose blood concentrations. Recipient POR*28 and CYP3A5 genotype were determined. RESULTS CYP3A5 expressers carrying at least 1 POR*28 allele had on average 18.3% lower tacrolimus predose concentrations and 20.2% lower concentration/dose ratios compared with CYP3A5 expressers with POR*1/*1 genotype (P = 0.002 and P = 0.001, respectively). In CYP3A5 nonexpressers, tacrolimus disposition did not significantly differ between POR genotypes. CONCLUSIONS In this small cohort of pediatric kidney transplant recipients, POR*28 genotype seems to explain part of the variability found in tacrolimus disposition, in addition to age and CYP3A5 genotype. This finding should be validated in a larger population, and it would be worthwhile to evaluate the clinical impact of this genotype.
Collapse
|
8
|
Pharmacogenomic Biomarker Information in FDA-approved Paediatric Drug Labels. Basic Clin Pharmacol Toxicol 2014; 116:438-44. [DOI: 10.1111/bcpt.12341] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/06/2014] [Indexed: 12/27/2022]
|
9
|
Stevens A, De Leonibus C, Hanson D, Whatmore A, Murray P, Donn R, Meyer S, Chatelain P, Clayton P. Pediatric perspective on pharmacogenomics. Pharmacogenomics 2014; 14:1889-905. [PMID: 24236488 DOI: 10.2217/pgs.13.193] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The advances in high-throughput genomic technologies have improved the understanding of disease pathophysiology and have allowed a better characterization of drug response and toxicity based on individual genetic make up. Pharmacogenomics is being recognized as a valid approach used to identify patients who are more likely to respond to medication, or those in whom there is a high probability of developing severe adverse drug reactions. An increasing number of pharmacogenomic studies are being published, most include only adults. A few studies have shown the impact of pharmacogenomics in pediatrics, highlighting a key difference between children and adults, which is the contribution of developmental changes to therapeutic responses across different age groups. This review focuses on pharmacogenomic research in pediatrics, providing examples from common pediatric conditions and emphasizing their developmental context.
Collapse
Affiliation(s)
- Adam Stevens
- Institute of Human Development, Medical & Human Sciences, University of Manchester & Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, 5th Floor Research, Oxford Road, Manchester, M13 9WL, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Gijsen VMGJ, van Schaik RH, Elens L, Soldin OP, Soldin SJ, Koren G, de Wildt SN. CYP3A4*22 and CYP3A combined genotypes both correlate with tacrolimus disposition in pediatric heart transplant recipients. Pharmacogenomics 2014; 14:1027-36. [PMID: 23837477 DOI: 10.2217/pgs.13.80] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Tacrolimus metabolism depends on CYP3A4 and CYP3A5. We aimed to determine the relationship between the CYP3A4*22 polymorphism and combined CYP3A genotypes with tacrolimus disposition in pediatric heart transplant recipients. METHODS Sixty pediatric heart transplant recipients were included. Tacrolimus doses and trough concentrations were collected in the first 14 days post-transplantation. CYP3A phenotypes were defined as extensive (CYP3A5*1 + CYP3A4*1/*1 carriers), intermediate (CYP3A5*3/*3 + CYP3A4*1/*1 carriers) or poor (CYP3A5*3/*3 + CYP3A4*22 carriers) metabolizers. RESULTS CYP3A4*22 carriers needed 30% less tacrolimus (p = 0.016) to reach similar target concentrations compared with CYP3A4*1/*1 (n = 56) carriers. Poor CYP3A metabolizers required 17% (p = 0.023) less tacrolimus than intermediate and 48% less (p < 0.0001) than extensive metabolizers. Poor metabolizers showed 18% higher dose-adjusted concentrations than intermediate (p = 0.35) and 193% higher than extensive metabolizers (p < 0.0001). CONCLUSION Analysis of CYP3A4*22, either alone or in combination with CYP3A5*3, may help towards individualization of tacrolimus therapy in pediatric heart transplant patients.
Collapse
Affiliation(s)
- Violette M G J Gijsen
- Erasmus MC Sophia Children's Hospital, Department of Pediatric Surgery & Intensive Care, Dr Molewaterplein 60, 3015 GJ Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
In the past several years, human genetics studies have progressed from monogenic to complex and common diseases because of the advancement in technologies. There is increased knowledge of the pharmacokinetics and pharmacogenomics of the drugs in adults as well as in children. These technological developments provided new diagnostic, prognostic, and therapeutic opportunities. We are now in a position to address many additional ambitious questions. For instance, in clinical medicine, interindividual variation in drug response is a major problem. Some of the heterogeneity of drug safety and efficacy among individuals can be explained by pharmacogenomics. It has also the potential to improve the treatment in both adults and children. In pediatrics however, there is ontogeny and metabolic capacity in children is different compared to adults. Several specific developmental changes may underlie some of the variability in drug response seen in children. They may also be responsible for adverse drug reactions (ADRs). Therefore, much of the diversity in drug effects cannot be explained by studying the genomic diversity alone. It is necessary to include the effect of growth (involves variations in gene expression) along with genetic differences when explaining the variability in treatment response. In this respect epigenomics may expand the scope of pharmacogenomics towards optimization of drug therapy. Future studies must focus on periods of maturation of the drug-metabolizing enzymes and polymorphisms in their genes by using candidate gene approach, gene expression analysis, genome-wide haplotype mapping, and proteomics. The integration of genetic data and clinical phenotypes along with the role of other factors is necessary to evaluate both efficacy and ADRs of any drug. It may require extensive genetic epidemiological studies spanning over many years.
Collapse
|
12
|
Madani S, Kauffman R, Simpson P, Lehr VT, Lai ML, Sarniak A, Tolia V. Pharmacokinetics and pharmacodynamics of famotidine and ranitidine in critically ill children. J Clin Pharmacol 2013; 54:201-5. [PMID: 24258773 DOI: 10.1002/jcph.219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/24/2013] [Indexed: 12/27/2022]
Abstract
To characterize and compare acid suppression (pharmacodynamics) and pharmacokinetics of IV famotidine and ranitidine in critically ill children at risk for stress gastritis. Single-blind, randomized study in PICU patients 6 months to 18 years requiring mechanical ventilation with continuous gastric pH monitoring, randomized to IV famotidine 12 mg/m(2) or ranitidine 60 mg/m(2) when gastric pH < 4.0 >1 hour with serial blood sampling following first dose. Twenty-four children randomized to either famotidine (n = 12) or ranitidine (n = 12). Sixteen out of twenty-four completed both PK and PD study arms (7/12 famotidine; 4.7 ± 3.4 years; 9/12 ranitidine; 6.6 ± 4.7 years; p = 0.38). Time to gastric pH 4.0 and total time pH above 4.0 similar with no difference in pH at 6 and 12 hours (p > 0.2). No difference between drugs in clearance, volume of distribution and half-life (p > 0.05). Ratio of AUC pH to AUC drug concentration 0-12 hours after first dose was significantly greater for famotidine (0.06849 ± 0.01460 SD) than ranitidine (0.02453 ± 0.01448; p < 0.001) demonstrating greater potency of famotidine. pH lowering efficacy of both drugs is similar. Greater potency of famotidine may offer clinical advantage due to lower drug exposure and less frequent dosing to achieve same pH lowering effect.
Collapse
Affiliation(s)
- Shailender Madani
- Division of Gastroenterology, Children's Hospital of Michigan, Wayne State University School of Medicine, Detroit, MI, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Comparative epidemiology of hospital-acquired adverse drug reactions in adults and children and their impact on cost and hospital stay--a systematic review. Eur J Clin Pharmacol 2013; 69:1985-96. [PMID: 23955174 DOI: 10.1007/s00228-013-1563-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/10/2013] [Indexed: 02/03/2023]
Abstract
PURPOSE To study and analyze the comparative impact of hospital-acquired adverse drug reactions (ADRs) in adult and pediatric patients in terms of the economic implications, (length of) hospital stay, and salient features in relation to the incidence rate, severity, morbidity, mortality, and preventability of the ADRs. METHODS A systematic search to identify and retrieve relevant articles/studies in the PubMed, Medline, Scopus, MEDPAR, and Cochrane databases and by the Google search engine was performed for the study period 2000 to April 2013. In total, 51 studies were identified on patients hospitalized for ADRs, and these were included in the study. The incidence rate of ADRs, their severity, mortality, morbidity, preventability, cost, and association with extended hospital stay due to ADRs were extracted and scrutinized. RESULTS Hospital-acquired ADRs are more widely studied in adults than in children, and the incidence rate is higher in the former. However, a wide variation in the incidence rate worldwide is observed in both groups. Irrespective of the ages of patients, ADRs are among the most frequent causes of morbidity and mortality. Interestingly, preventable ADRs are more frequently observed in patients at the younger and older ends of the age spectrum. Hospital-acquired ADRs place an immense economic burden on healthcare systems, with the overall cost for a hospitalized patient with an ADR reported to be $2,401 per patient, which is equivalent to a 19.86 % additional increase in the total cost of care and an increase in average length of hospital stay of 8.25 %. CONCLUSION Based on the findings of this review, we suggest that excellent assertive measures of pharmacovigilance with the aim to diminish the incidence rate of hospital-acquired ADRs and support the development of interventions are needed to promote vital facets of drug safety with an overall objective to avert potential ADRs.
Collapse
|
14
|
Du W, Tutag Lehr V, Caverly M, Kelm L, Reeves J, Lieh-Lai M. Incidence and Costs of Adverse Drug Reactions in a Tertiary Care Pediatric Intensive Care Unit. J Clin Pharmacol 2013; 53:567-73. [DOI: 10.1002/jcph.75] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/14/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Wei Du
- Department of Pediatrics, School of Medicine; Wayne State University; Detroit, MI; USA
| | - Victoria Tutag Lehr
- Pharmacy Practice, Eugene Applebaum College of Pharmacy & Health Sciences; Wayne State University; Detroit, MI; USA
| | - Mary Caverly
- Critical Care Medicine; Children's Hospital of Michigan; Detroit, MI; USA
| | - Lauren Kelm
- Critical Care Medicine; Children's Hospital of Michigan; Detroit, MI; USA
| | - Jaxk Reeves
- Department of Statistics; University of Georgia; Athens, GA; USA
| | - Mary Lieh-Lai
- Department of Pediatrics, School of Medicine; Wayne State University; Detroit, MI; USA
| |
Collapse
|
15
|
Du W, Lehr VT, Lieh-Lai M, Koo W, Ward RM, Rieder MJ, Van Den Anker JN, Reeves JH, Mathew M, Lulic-Botica M, Aranda JV. An algorithm to detect adverse drug reactions in the neonatal intensive care unit. J Clin Pharmacol 2013; 53:87-95. [PMID: 23400748 DOI: 10.1177/0091270011433327] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 11/17/2011] [Indexed: 01/22/2023]
Abstract
Critically ill newborns in neonatal intensive care units (NICUs) are at greater risk of developing adverse drug reactions (ADRs). Differentiation of ADRs from reactions associated with organ dysfunction/immaturity is difficult. Current ADR algorithm scoring was established arbitrarily without validation in infants. The study objective was to develop a valid and reliable algorithm to identify ADRs in the NICU. Algorithm development began with a 24-item questionnaire for data collection on 100 previously suspected ADRs. Five pediatric pharmacologists independently rated cases as definite, probable, possible, and unlikely ADRs. Consensus "gold standard" was reached via teleconference. Logistic regression and iterative C programs were used to derive the scoring system. For validation, 50 prospectively collected ADR cases were assessed by 3 clinicians using the new algorithm and the Naranjo algorithm. Weighted kappa and intraclass correlation coefficient (ICC) were used to compare validity and reliability of algorithms. The new algorithm consists of 13 items. Kappa and ICC of the new algorithm were 0.76 and 0.62 versus 0.31 and 0.43 for the Naranjo algorithm. The new algorithm developed using actual patient data is more valid and reliable than the Naranjo algorithm for identifying ADRs in the NICU population. Because of the relatively small and nonrandom samples, further refinement and additional testing are needed.
Collapse
Affiliation(s)
- Wei Du
- Department of Pediatrics, School of Medicine, Wayne State University, Detroit, MI, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Khan LM, Al-Harthi SE, Saadah OI. Adverse drug reactions in hospitalized pediatric patients of Saudi Arabian University Hospital and impact of pharmacovigilance in reporting ADR. Saudi Pharm J 2012; 21:261-6. [PMID: 23960842 DOI: 10.1016/j.jsps.2012.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 09/11/2012] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Children are more vulnerable to ADRs, and this susceptibility is compounded due to hospitalization. There is a lack of local data regarding the potential risk of ADRs in hospitalized pediatric patients. Therefore, this study is designed to identify the frequent nature, severity of adverse drug reactions, drugs implicated and factors influencing ADRs. METHODS Intensive monitoring study of ADRs was done in hospitalized pediatric patients of King Abdulaziz University Hospital, Jeddah from January to December 2011, with an analogous retrospective study for the preceding year to determine incidence rate, demographic aspects, causality appraisal, polypharmacy, body organs/systems involved and drugs implicated in ADR. Comparison of the two data was done to determine the impact of pharmacovigilance. RESULTS Incidence rate of ADRs in retrospective study was (4.50%) and (8.2%) in prospective study. ADR was more in patients who received 5-6 drugs, which was (15.5%) in retrospective study and (22.1%) in prospective study. Regarding age, it was the highest in patients of 0-1 year of age which was (40.7%) in retrospective study and (38.8%) in prospective study. Anti-infective agents were the most frequently involved in ADR (40.8%) in prospective study and (48.2%) and retrospective study. This study also demonstrated that, there was high susceptibility of the skin to the ADR which was (37%) in retrospective study and (42.9%) in prospective study. None of the ADRs proved to be fatal. CONCLUSION Well premeditated intensive monitoring approach in pharmacovigilance amplifies the ADR detection, which can persuade healthcare providers into more drug safety.
Collapse
Affiliation(s)
- Lateef M Khan
- Department of Pharmacology, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | |
Collapse
|
17
|
Wagner J, Leeder JS. Pediatric pharmacogenomics: a systematic assessment of ontogeny and genetic variation to guide the design of statin studies in children. Pediatr Clin North Am 2012; 59:1017-37. [PMID: 23036242 PMCID: PMC5847265 DOI: 10.1016/j.pcl.2012.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The dose-exposure-response relationship for drugs may differ in pediatric patients compared with adults. Many clinical studies have established drug dose-exposure relationships across the pediatric age spectrum; however, genetic variation was seldom included. This article applies a systematic approach to determine the relative contribution of development and genetic variation on drug disposition and response using HMG-CoA reductase inhibitors as a model. Application of the approach drives the collection of information relevant to understanding the potential contribution of ontogeny and genetic variation to statin dose-exposure-response in children, and identifies important knowledge deficits to be addressed through the design of future studies.
Collapse
Affiliation(s)
- Jonathan Wagner
- Section of Cardiology, Children's Mercy Hospital and Clinics, University of Missouri-Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO 64108, USA.
| | - J. Steven Leeder
- Division of Clinical Pharmacology and Medical Toxicology, Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Children’s Mercy Hospital and Clinics, 2401 Gillham Road, Kansas City, Missouri 64108
| |
Collapse
|
18
|
Wall CA, Croarkin PE, Swintak C, Koplin BA. Psychiatric pharmacogenomics in pediatric psychopharmacology. Child Adolesc Psychiatr Clin N Am 2012; 21:773-88. [PMID: 23040901 DOI: 10.1016/j.chc.2012.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This article provides an overview of where psychiatric pharmacogenomic testing stands as an emerging clinical tool in modern psychotropic prescribing practice, specifically in the pediatric population. This practical discussion is organized around the state of psychiatric pharmacogenomics research when choosing psychopharmacologic interventions in the most commonly encountered mental illnesses in youth. As with the rest of the topics on psychopharmacology for children and adolescents in this publication, a clinical vignette is presented, this one highlighting a clinical case of a 16 year old genotyped during hospitalization for recalcitrant depression.
Collapse
Affiliation(s)
- Christopher A Wall
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
19
|
Sears K, Ross-White A, Godfrey CM. The incidence, prevalence and contributing factors associated with the occurrence of medication errors for children and adults in the community setting: a systematic review. ACTA ACUST UNITED AC 2012; 10:2350-2464. [DOI: 10.11124/jbisrir-2012-35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
20
|
Shaw K, Amstutz U, Carleton BC. Using pharmacogenetics to understand adverse drug reactions in children. Paediatr Child Health 2011; 16:537-8. [PMID: 23115490 PMCID: PMC3223886 DOI: 10.1093/pch/16.9.537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2011] [Indexed: 11/14/2022] Open
Affiliation(s)
- Kaitlyn Shaw
- Faculty of Medicine, Department of Paediatrics, University of British Columbia
- Pharmaceutical Outcomes Programme, BC Children’s Hospital
- Child & Family Research Institute, Vancouver, British Columbia
| | - Ursula Amstutz
- Faculty of Medicine, Department of Paediatrics, University of British Columbia
- Pharmaceutical Outcomes Programme, BC Children’s Hospital
- Child & Family Research Institute, Vancouver, British Columbia
| | - Bruce C Carleton
- Faculty of Medicine, Department of Paediatrics, University of British Columbia
- Pharmaceutical Outcomes Programme, BC Children’s Hospital
- Child & Family Research Institute, Vancouver, British Columbia
| |
Collapse
|
21
|
Piana C, Surh L, Furst-Recktenwald S, Iolascon A, Jacqz-Aigrain EM, Jonker I, Russo R, van Schaik RHN, Wessels J, Della Pasqua OE. Integration of pharmacogenetics and pharmacogenomics in drug development: implications for regulatory and medical decision making in pediatric diseases. J Clin Pharmacol 2011; 52:704-16. [PMID: 21566202 DOI: 10.1177/0091270011401619] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This article aims to provide an overview of the current situation regarding pharmacogenetic and pharmacogenomic (PG) studies in pediatrics, with a special focus on the role of PG data in the regulatory decision-making process. Despite the gap in pharmacogenetic research due to the lack of translational studies in adults and children, several technologies exist in drug development and biomarkers validation, which could supply valuable information concerning labeling and dosing recommendations. If performed under strict good clinical practice quality criteria, such findings could be included in the submission package of new chemical entities and used as additional information for prescribers, supporting further evaluation and understanding of the efficacy and safety profile of new medicines. Even though regulatory authorities may be aware of the potential role of PG in medical practice and guidances are available about the integration of PG in drug development, most data obtained from PG studies are not used by prescribers. The challenge is to better understand whether PG markers can be used to assess potential differences in drug response during the clinical program, so PG data can be integrated into the regulatory decision-making process, enabling the introduction of labeling information that promotes optimal dosing in the pediatric population.
Collapse
Affiliation(s)
- Chiara Piana
- Division of Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Understanding the pharmacokinetics and pharmacodynamics of drugs used in psychopharmacology across the pediatric age spectrum from infants to adolescents represents a major challenge for clinicians. In pediatrics, treatment protocols use either standard dose reductions for these drugs for children below a certain age or use less conventional parameters such as weight for allometric dosing. The rationale behind this, however, is often lacking. In this review current available data on the developmental changes in absorption, distribution, metabolism, and elimination of drugs are presented with a specific focus on metabolic pathways, indicating significant differences in the development of enzymes involved in the biotransformation of drugs used in psychopharmacology. Major emphasis will be given to the genetic variation in CYP2D6 activity, the most important enzyme for the metabolism of many psychotropic medications. Finally, this review will summarize the role of the developmental pharmacologist in ensuring rational use of drugs in children with developmental disabilities and in translating pharmacological concepts from the bench to the clinic.
Collapse
Affiliation(s)
- Johannes N van den Anker
- Division of Pediatric Clinical Pharmacology, George Washington University Medical Center, Children's National Medical Center, Washington DC, USA.
| |
Collapse
|
23
|
Abstract
The advances in developmental pharmacokinetics during the past decade reside with an enhanced understanding of the influence of growth and development on drug absorption, distribution, metabolism, and excretion (ADME). However, significant information gaps remain with respect to our ability to characterize the impact of ontogeny on the activity of important drug metabolizing enzymes, transporters, and other targets. The ultimate goal of rational drug therapy in neonates, infants, children, and adolescents resides with the ability to individualize it based on known developmental differences in drug disposition and action. The clinical challenge in achieving this is accounting for the variability in all of the contravening factors that influence pharmacokinetics and pharmacodynamics (e.g., genetic variants of ADME genes, different disease phenotypes, disease progression, and concomitant treatment). Application of novel technologies in the fields of pharmacometrics (e.g., in silico simulation of exposure-response relationships; disease progression modeling), pharmacogenomics and biomarker development (e.g., creation of pharmacodynamic surrogate endpoints suitable for pediatric use) are increasingly making integrated approaches for developmentally appropriate dose regimen selection possible.
Collapse
Affiliation(s)
- Johannes N van den Anker
- Division of Pediatric Clinical Pharmacology, Department of Pediatrics, Children's National Medical Center, NW, Washington, DC 20010, USA.
| | | | | |
Collapse
|
24
|
Russo R, Capasso M, Paolucci P, Iolascon A. Pediatric pharmacogenetic and pharmacogenomic studies: the current state and future perspectives. Eur J Clin Pharmacol 2010; 67 Suppl 1:17-27. [PMID: 21069522 DOI: 10.1007/s00228-010-0931-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 10/16/2010] [Indexed: 11/26/2022]
Abstract
Genetic differences among individuals can explain some of the variability observed during drug treatment. Many studies have correlated the different pharmacological response to genetic variability, but most of them have been conducted on adult populations. Much less attention has been given to the pediatric population. Pediatric patients constitute a vulnerable group with regard to rational drug prescribing since they present differences arising from the various stages of development. However, only a few steps have been made in developmental pharmacogenomics. This review attempts to describe the current methods for pharmacogenetic and pharmacogenomic studies, providing some of the most studied examples in pediatric patients. It also gives an overview on the implication and importance of microRNA polymorphisms, transcriptomics, metabonomics, and proteomics in pharmacogenetics and pharmacogenomics studies.
Collapse
Affiliation(s)
- Roberta Russo
- Department of Biochemistry and Medical Biotechnologies, University Federico II of Naples and CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy.
| | | | | | | |
Collapse
|
25
|
Loo TT, Ross CJD, Sistonen J, Visscher H, Madadi P, Koren G, Hayden MR, Carleton BC. Pharmacogenomics and active surveillance for serious adverse drug reactions in children. Pharmacogenomics 2010; 11:1269-85. [PMID: 20860467 DOI: 10.2217/pgs.10.111] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Juxtaposing clinical pharmacology with human genetics, pharmacogenomics utilizes a patient’s genetic information to identify genetic variants that have the potential to provide clinically relevant predictions of toxicity and efficacy. The goal is to develop personalized and genetic-based predictions of an individual’s drug response and likelihood of experiencing an adverse drug reaction. The Canadian Pharmacogenomics Network for Drug Safety (CPNDS) has implemented active adverse drug reaction surveillance to monitor and discover genetic markers related to serious adverse drug reactions in the pediatric population. Evidence-based pharmacogenomics research will inform public policy and influence drug benefit–risk decision-making. Regulatory processes and future challenges in pharmacogenomics research will be discussed.
Collapse
Affiliation(s)
- Tenneille T Loo
- Faculty of Medicine, Department of Paediatrics, UBC, Vancouver, Canada
- Pharmaceutical Outcomes Programme, BC Children’s Hospital, Vancouver, Canada
- Child & Family Research Institute, Vancouver, Canada
| | - Colin JD Ross
- Centre for Molecular Medicine & Therapeutics, UBC, Vancouver, Canada
- Faculty of Medicine, Department of Medical Genetics, UBC, Vancouver, Canada
| | - Johanna Sistonen
- Centre for Molecular Medicine & Therapeutics, UBC, Vancouver, Canada
- Faculty of Medicine, Department of Medical Genetics, UBC, Vancouver, Canada
| | - Henk Visscher
- Centre for Molecular Medicine & Therapeutics, UBC, Vancouver, Canada
- Faculty of Medicine, Department of Medical Genetics, UBC, Vancouver, Canada
| | - Parvaz Madadi
- Division of Clinical Pharmacology & Toxicology, Hospital for Sick Children, Toronto, Canada
- Motherisk Program, Hospital for Sick Children, Toronto, Canada
| | - Gideon Koren
- Motherisk Program, Hospital for Sick Children, Toronto, Canada
- Department of Physiology & Pharmacology, University of Western Ontario (UWO), Ontario, Canada
- Schulich School of Medicine & Dentistry, UWO, Ontario, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine & Therapeutics, UBC, Vancouver, Canada
- Faculty of Medicine, Department of Medical Genetics, UBC, Vancouver, Canada
| | | |
Collapse
|
26
|
Abstract
Pharmacogenomics, the study of specific genetic variations and their effect on drug response, will likely give rise to many applications in maternal-fetal and neonatal medicine; yet, an understanding of these applications in the field of obstetrics and gynecology and neonatal pediatrics is not widespread. This review describes the underpinnings of the field of pharmacogenomics and summarizes the current pharmacogenomic inquiries in relation to maternal-fetal medicine-including studies on various fetal and neonatal genetic cytochrome P450 (CYP) enzyme variants and their role in drug toxicities (for example, codeine metabolism, sepsis and selective serotonin reuptake inhibitor (SSRI) toxicity). Potential future directions, including alternative drug classification, improvements in drug efficacy and non-invasive pharmacogenomic testing, will also be explored.
Collapse
|
27
|
Leeder JS, Kearns GL, Spielberg SP, van den Anker J. Understanding the relative roles of pharmacogenetics and ontogeny in pediatric drug development and regulatory science. J Clin Pharmacol 2010; 50:1377-87. [PMID: 20150527 DOI: 10.1177/0091270009360533] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Understanding the dose-exposure-response relationship across the pediatric age spectrum from preterm and term newborns to infants, children, adolescents, and adults is a major challenge for clinicians, pharmaceutical companies, and regulatory agencies. Over the past 3 decades, clinical investigations of many drugs commonly used in pediatric therapeutics have provided valuable insights into age-associated differences in drug disposition and action. However, our understanding of the contribution of genetic variation to variability in drug disposition and response in children generally has lagged behind that of adults. This article proposes a systematic approach that can be used to assess the relative contributions of ontogeny and genetic variation for a given compound. Application of the strategy is illustrated using the current regulatory dilemma posed by the safety and effectiveness of over-the-counter cough and cold remedies as an example. The results of the analysis can be used to aid in the design of studies to yield maximally informative data in pediatric populations of different ages and developmental stages and thereby improve the efficiency of study design.
Collapse
Affiliation(s)
- J Steven Leeder
- Division of Clinical Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, School of Medicine, University of Missouri-Kansas City, 2401 Gillham Road, Kansas City, MO 64108, USA.
| | | | | | | |
Collapse
|
28
|
Carleton BC, Poole RL, Smith MA, Leeder JS, Ghannadan R, Ross CJD, Phillips MS, Hayden MR. Adverse drug reaction active surveillance: developing a national network in Canada's children's hospitals. Pharmacoepidemiol Drug Saf 2009; 18:713-21. [DOI: 10.1002/pds.1772] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
29
|
Avard D, Silverstein T, Sillon G, Joly Y. Researchers' perceptions of the ethical implications of pharmacogenomics research with children. Public Health Genomics 2009; 12:191-201. [PMID: 19204423 PMCID: PMC2790793 DOI: 10.1159/000189633] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This paper presents the results of an exploratory qualitative study that assesses Canadian pediatric researchers' perceptions of a pre-selected group of ethical issues raised by pharmacogenomics research with children. METHODS As a pilot study, we conducted semi-structured telephone interviews with Canadian pediatric pharmacogenomic researchers. The interviews were guided by the following themes: (1) benefits and risks of inclusion, (2) the consent/assent process, and (3) the return of research results. RESULTS Issues about assent, consent, risks and benefits, as well as the communication of results were addressed by the respondents. Some issues, such as the unique vulnerability of children, the long term privacy concerns associated with biobanking, additional core elements that need to be discussed and included in the consent/assent forms, as well as the challenges of communicating research results in a pediatric research were not explicitly identified by the respondents. CONCLUSION Further consideration should be given to address the ethical challenges of including children in pharmacogenomics research. This exploratory study indicates that further guidance is needed if children are to be protected and yet benefit from such research.
Collapse
Affiliation(s)
- D Avard
- Law Faculty, Centre de Recherche en Droit Public, Université de Montréal, Montréal, Qué, Canada.
| | | | | | | |
Collapse
|
30
|
Adjei AA, Gaedigk A, Simon SD, Weinshilboum RM, Leeder JS. Interindividual variability in acetaminophen sulfation by human fetal liver: implications for pharmacogenetic investigations of drug-induced birth defects. ACTA ACUST UNITED AC 2008; 82:155-65. [PMID: 18232020 DOI: 10.1002/bdra.20535] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Acetaminophen (APAP) use in early pregnancy has been associated with the risk of gastroschisis, a rare but serious congenital defect of the abdominal wall. The purpose of this study was to characterize the variability of APAP sulfation in a panel of human fetal livers and to identify the sulfotransferases (SULT) isoform(s) responsible for catalyzing that activity. METHODS APAP sulfation was determined in a panel of human fetal (n = 73) and postnatal (n = 18) liver cytosol preparations and correlated with the catalytic activity of various SULT isoforms as determined using prototypic substrates and specific antibodies. RESULTS Of 10 heterologously expressed SULT isoforms examined, SULT1A1, SULT1A3/4, SULT1E1, and SULT2A1 all catalyzed the formation of APAP sulfate with K(m) values of 2.4, 1.5, 1.9, and 3.7 mM, respectively. Catalytic activities for these four isoforms were expressed at varying levels in human fetal liver, and APAP sulfation was positively correlated with each of the four prototypic activities. Several regression and clustering approaches revealed that SULT1A3/4 was the primary determinant of prenatal APAP sulfation but that SULT1A1 or SULT1E1 were also major contributors in subsets of samples. CONCLUSIONS The results of this study lead to the hypothesis that genetic variation in SULT1A3/4 represents a risk factor for the development of gastroschisis in the offspring of mothers exposed to APAP early in pregnancy. Interpretation of genetic association studies conducted to test this hypothesis will be complicated by the variable contributions of other SULTs toward APAP-sulfate formation in individual subjects.
Collapse
Affiliation(s)
- Araba A Adjei
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
31
|
Husain A, Loehle JA, Hein DW. Clinical pharmacogenetics in pediatric patients. Pharmacogenomics 2008; 8:1403-11. [PMID: 17979513 DOI: 10.2217/14622416.8.10.1403] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pediatric pharmacogenetic studies have the potential to improve the quality of medical care for children. The pediatric population presents a unique pharmacogenetic challenge as children have the additional complexity of ontological phenotypes that impact their drug response. Prescribing medications in children has historically been largely empirical, but utilization of pharmacogenetic information will allow pediatricians to gain key information regarding which patients are best suited for a particular therapeutic agent and which patients may be at risk for serious potentially life-threatening complications from standard treatment regimens. Although large, prospective, multisite investigators are still needed, we illustrate selective clinical examples of the pharmacogenetics for treatment of transplantation, asthma, leukemia and attention-deficit hyperactivity disorder in pediatric patients.
Collapse
Affiliation(s)
- Anwar Husain
- University of Louisville, Department of Pharmacology & Toxicology and James Graham Brown Center, School of Medicine, Louisville, KY, USA
| | | | | |
Collapse
|
32
|
Ross CJD, Carleton B, Warn DG, Stenton SB, Rassekh SR, Hayden MR. Genotypic Approaches to Therapy in Children: A National Active Surveillance Network (GATC) to Study the Pharmacogenomics of Severe Adverse Drug Reactions in Children. Ann N Y Acad Sci 2007; 1110:177-92. [PMID: 17911433 DOI: 10.1196/annals.1423.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A striking failure of modern medicine is the debilitating and lethal consequences of adverse drug reactions (ADRs), which rank as one of the top 10 leading causes of death and illness in the developed world with direct medical costs of 137-177 billion annually US dollars in the USA. Although many factors influence the effect of medications (e.g., age, organ function, drug interactions), genetic factors account for 20% to 95% of drug response variability and play a significant role in the incidence and severity of ADRs. The field of pharmacogenomics seeks to identify genetic factors responsible for individual differences in drug efficacy and ADRs. Pharmacogenomics has led to several genetic tests that provide clinical dosing recommendations. The Genetic Approaches to Therapy in Children (GATC) project is a national project established in Canada to identify novel predictive genomic markers for severe ADRs in children. An ADR surveillance network has been established in eight of Canada's major children's hospitals, serving up to 75% of all Canadian children. The goal of the project is to identify patients experiencing specific ADRs and matched controls, collect DNA samples, and apply genomics-based technologies to identify ADR-associated genetic markers with the goal of preventing serious ADRs in susceptible children.
Collapse
Affiliation(s)
- Colin J D Ross
- Department of Medical Genetics, University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Krekels EHJ, van den Anker JN, Baiardi P, Cella M, Cheng KY, Gibb DM, Green H, Iolascon A, Jacqz-Aigrain EM, Knibbe CAJ, Santen GWE, van Schaik RHN, Tibboel D, Della Pasqua OE. Pharmacogenetics and paediatric drug development: issues and consequences to labelling and dosing recommendations. Expert Opin Pharmacother 2007; 8:1787-99. [PMID: 17696784 DOI: 10.1517/14656566.8.12.1787] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The area of pharmacogenetics (PGt) is evolving rapidly. However, ongoing efforts in this field are not aligned with the requirements for the inclusion of clinically relevant findings into the label, especially with reference to paediatric indications. Clinical research in children poses unique issues from a practical and technical perspective, but many challenges can be overcome by applying advanced study design and data analysis methods. When investigating the role of PGt factors on treatment effect, all features that influence drug response must be taken into account. Yet, PGt often has a privileged status in research protocols, with PGt factors evaluated independently from other determinants of response, instead of being regarded as other demographic or clinical covariates (e.g., age, renal function). At present, guidelines to incorporate PGt findings into label statements are lacking in part because this is a new and incompletely understood area. This situation is no longer acceptable. To achieve the potential that PGt can offer to drug development and ultimately to drug prescription, academia, industry and regulatory agencies need to pool resources on the revision of study design and data analysis requisites, bringing in model-based methodologies to enable accurate interpretation of results and provide appropriate labelling recommendations.
Collapse
Affiliation(s)
- Elke H J Krekels
- Clinical Pharmacology & Discovery Medicine, GlaxoSmithKline, Greenford, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ross CJD, Katzov H, Carleton B, Hayden MR. Pharmacogenomics and its implications for autoimmune disease. J Autoimmun 2007; 28:122-8. [PMID: 17418528 DOI: 10.1016/j.jaut.2007.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A striking failure of modern medicine is the debilitating and lethal consequences of adverse drug reactions (ADRs) which rank as one of the top ten leading causes of death and illness in the developed world with direct medical costs of 137-177 billion US dollars annually in the USA. Although many factors influence the effect of medications (i.e. age, organ function, drug interactions), genetic factors account for 20-95% of drug response variability and play a significant role in the incidence and severity of ADRs. The field of pharmacogenomics seeks to identify genetic factors responsible for individual differences in drug efficacy and adverse drug reactions. Pharmacogenomics has led to several genetic tests that provide clinical dosing recommendations. For autoimmune disease, pharmacogenomics has led to several DNA-based tests to improve drug selection, optimize dosing, and minimize the risk of toxicity. The 'GATC' project is a nation-wide project established in Canada to identify novel predictive genomic markers of severe ADRs in children. An ADR surveillance network has been established in all of Canada's major children's hospitals, serving up to 75% of all Canadian children. The goal of the project is to identify patients experiencing specific ADRs, collect DNA samples, and apply genomics-based technologies to identify ADR-associated genetic markers.
Collapse
Affiliation(s)
- Colin J D Ross
- Department of Medical Genetics, University of British Columbia (UBC), Centre for Molecular Medicine and Therapeutics, 950 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | | | | | | |
Collapse
|
35
|
Blake MJ, Castro L, Leeder JS, Kearns GL. Ontogeny of drug metabolizing enzymes in the neonate. Semin Fetal Neonatal Med 2005; 10:123-38. [PMID: 15701578 DOI: 10.1016/j.siny.2004.11.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Fetal exposure to xenobiotics is modulated to a considerable degree by the metabolic capabilities of the mother and the placenta. However, once liberated from the uterine environment the neonate is instantly exposed to a wide array of new macromolecules in the form of byproducts of cellular metabolism, dietary constituents, environmental toxins and pharmacologic agents. The rapid and efficient biotransformation of these compounds by Phase I and Phase II drug-metabolizing enzymes is an essential process if the infant is to avoid the accumulation of reactive compounds that could produce cellular injury or tissue dysfunction. Genetic polymorphisms and environmental factors are known to contribute dramatically to individual variation in the activity of drug-metabolizing enzymes. More recently, it has become apparent that programmed, developmental, regulatory events occur - independent of genotype - which further add to individual variation in drug metabolism. An appreciation of the impact of ontogeny on the expression and functional activity of the major drug-metabolizing enzymes enables the practicing clinician to predict the ultimate consequence of drug administration in the neonate to help guide optimal drug therapy.
Collapse
Affiliation(s)
- Michael J Blake
- Department of Pediatrics, University of Missouri - Kansas City, Division of Pediatric Pharmacology and Medical Toxicology, The Children's Mercy Hospitals and Clinics, 2401 Gillham Road, Kansas City, MO 64108, USA
| | | | | | | |
Collapse
|