1
|
Ettienne EB, Grant-Kels JM, Striano P, Russo E, Neubauer D, Rose K. Pharmacogenomics and pediatric drug development: science and political power. A narrative review. Expert Opin Pharmacother 2024; 25:2367-2373. [PMID: 39268964 DOI: 10.1080/14656566.2024.2401429] [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] [Received: 06/24/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
INTRODUCTION Pharmacogenomics (PGx) investigates how genomes control enzyme expression. Developmental pharmacology (DP) describes the temporal sequence of enzymes impacting absorption, distribution, metabolism, and excretion (ADME) of food and drugs. AREAS COVERED US and European Union (EU) legislation facilitate and/or enforce pediatric studies for all new drugs, called overall 'pediatric drug development' (PDD). DP and PDD look at patients' chronological age, but oscillate between legal and physiological meanings of the term 'child.' Children's bodies become mature with puberty. EXPERT OPINION Decades after first DP observations in babies, PGx offers a better understanding of the variability of safety and efficacy of drugs, of the process of aging, and of shifting enzyme patterns across aging. We should rethink and revise outdated interpretations of ADME changes in minors. The Declaration of Helsinki forbids pointless studies that some pediatric researchers and regulatory agencies, more so the EMA than the FDA, demand pointless pediatric studies is regrettable. Medicine needs to differentiate between legal and physiological meanings of the term 'child' and should use objective measures of maturity.
Collapse
Affiliation(s)
| | - Jane M Grant-Kels
- Dermatology, Pathology, and Pediatric Dermatology, University of Connecticut Health Center, Farmington, USA
| | | | - Emilio Russo
- Pharmacology, University of Magna Graecia, Catanzaro, Italy
| | - David Neubauer
- Department of Child, Adolescent & Developmental Neurology, University Childrens' Hospital, Ljubljana, Slovenia
| | - Klaus Rose
- klausrose Consulting, Riehen, Switzerland
| |
Collapse
|
2
|
Buendía JA, Salazar AFZ. Genotype-driven asthma prescribing of inhaled corticosteroids and long-acting β2-agonist: A cost-effectiveness analysis. Pediatr Pulmonol 2024; 59:2449-2456. [PMID: 38661231 DOI: 10.1002/ppul.27037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/17/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Predicting response to inhaled corticosteroids (ICSs) + long-acting β2-agonist (LABA) by previously detecting the presence of Arg16Gly ADRB2 genotype is a strategy that could reduce and optimize the management of asthmatic patients. There is a need for economic evaluations to facilitate the implementation of such tests. This research aims to evaluate the cost-effectiveness of Arg16Gly ADRB2 screening in children with asthma in Colombia. METHODS From the perspective of a third-party payer, we conducted a cost-effectiveness analysis to determine the cost and quality-adjusted life-years (QALYs) of genotype-driven asthma prescribing based on the Arg16Gly ADRB2 genotype versus current treatment based on no genetic testing. Using four state-transition models, we estimate cost and QALYs employing micro-simulation modeling with a time horizon of 10 years and a cycle length of 1 week. Cost-effectiveness was assessed at a willingness-to-pay (WTP) value of US$5180. RESULTS The mean incremental cost of strategy genetic testing versus no genetic testing is US$ -6809. The mean incremental benefit of strategy genetic testing is 16 QALYs. The incremental net monetary benefit of strategic genetic testing versus no genetic testing is US$ 88,893. Genetic testing is the strategy with the highest expected net benefit. The outcomes derived from our primary analysis remained robust when subjected to variations in all underlying assumptions and parameter values. CONCLUSION Genetic testing of Arg16Gly ADRB2 is a cost-effective strategy to address asthma management in asthmatic children requiring ICS+LABA. This result should encourage the generation of more evidence and the incorporation of such evidence into clinical practice guidelines for pediatric asthma.
Collapse
Affiliation(s)
- Jefferson Antonio Buendía
- Research Group in Pharmacology and Toxicology, Department of Pharmacology and Toxicology, University of Antioquia, Medellín, Colombia
- Warwick Evidence, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Andrés Felipe Zuluaga Salazar
- Research Group in Pharmacology and Toxicology, Department of Pharmacology and Toxicology, University of Antioquia, Medellín, Colombia
- Laboratorio Integrado de Medicina Especializada (LIME), Facultad de Medicina, IPS Universitaria, Universidad de Antioquia, Antioquia, Colombia
| |
Collapse
|
3
|
Sakon CM, Sales C, Mertami S, Raibulet A, Schulte RR, Slaven JE, Tillman EM. Utilization of supportive care medications and opportunities for pre-emptive pharmacogenomic testing in pediatric and young adults with leukemia. Pediatr Hematol Oncol 2024; 41:480-488. [PMID: 38904214 PMCID: PMC11410515 DOI: 10.1080/08880018.2024.2368007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
This study aimed to evaluate the utilization of drugs with pharmacogenomic guidelines (PGx-drugs) for personalized dosing in pediatric leukemia. A retrospective observational study of pediatric leukemia patients admitted between 2009-2019 at a single-center academic children's hospital was conducted to determine PGx-drug exposure within 3 years of diagnosis. Along with baseline demographic and clinical characteristics of these patients, data regarding dates of diagnosis, relapse, death were collected. During the study period, inclusion criteria were met by 714 patients. The most frequently given medications were ondansetron (96.1%), morphine (92.2%), and allopurinol (85.3%) during the study period. In this cohort, 82% of patients received five or more PGx-drugs. Patients diagnosed with acute myeloid leukemia and leukemia unspecified were prescribed more PGx-drugs than other types of leukemia. There was a significant relationship between age at diagnosis and the number of PGx-drugs prescribed. Adolescents and adults both received a median of 10 PGx-drugs, children received a median of 6 PGx-drugs, and infants received a median of 7 PGx-drugs (p < 0.001). Patients with recurrent leukemia had significantly more PGx-drugs prescribed compared to those without recurrent disease, 10 drugs and 6 drugs, respectively (p < 0.001). Patients diagnosed with childhood leukemia are high utilizers of PGx-drugs. There is a vital need to understand how PGx testing may be utilized to optimize treatment and enhance quality of life. Preemptive PGx testing is a tool that aids in optimization of drug therapy and decreases the need for later treatment modifications. This can result in financial savings from decreased health-care encounters.
Collapse
Affiliation(s)
| | - Carmina Sales
- Purdue University College of Pharmacy, Purdue University
| | - Selsbiel Mertami
- Division of Clinical Pharmacology, Indiana University School of Medicine (IUSM)
| | - Andra Raibulet
- College of Pharmacy and Health Sciences, Butler University
| | - Rachael R Schulte
- Department of Pediatrics, Division of Pediatric Hematology/Oncology/Stem Cell Transplant, IUSM
| | | | - Emma M. Tillman
- Division of Clinical Pharmacology, Indiana University School of Medicine (IUSM)
| |
Collapse
|
4
|
Gallaway KA, Cann K, Oetting K, Rothenberger M, Raibulet A, Slaven JE, Suhrie K, Tillman EM. The Potential Impact of Preemptive Pharmacogenetic Genotyping in the Neonatal Intensive Care Unit. J Pediatr 2023; 259:113489. [PMID: 37201679 DOI: 10.1016/j.jpeds.2023.113489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/26/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
OBJECTIVE To evaluate the use of drugs with pharmacogenomic (PGx) guidelines from the Clinical Pharmacogenetics Implementation Consortium in early childhood. STUDY DESIGN A retrospective observational study of patients admitted to the neonatal intensive care (NICU) between 2005 and 2018 with at least 1 subsequent hospitalization at or after 5 years of age was performed to determine PGx drug exposure. Data regarding hospitalizations, drug exposures, gestational age, birth weight, and congenital anomalies and/or a primary genetic diagnosis were collected. Incidence of PGx drug and drug class exposures was determined and patient specific factors predictive of exposure were investigated. RESULTS During the study, 19 195 patients received NICU care and 4196 (22%) met study inclusion; 67% received 1-2, 28% 3-4, and 5% 5 or more PGx-drugs in early childhood. Preterm gestation, low birth weight (<2500 g), and the presence of any congenital anomalies and/or a primary genetic diagnosis were statistically significant predictors of Clinical Pharmacogenetics Implementation Consortium drug exposures (P < .01, P < .01, P < .01, respectively). CONCLUSIONS Preemptive PGx testing in patients in the NICU could have a significant impact on medical management during the NICU stay and throughout early childhood.
Collapse
Affiliation(s)
- Katherine A Gallaway
- Division of Pediatric Critical Care, Indiana University School of Medicine, Indianapolis, IN
| | - Kayla Cann
- Purdue University College of Pharmacy, Purdue University, West Lafayette, IN
| | - Katherine Oetting
- Purdue University College of Pharmacy, Purdue University, West Lafayette, IN
| | - Mary Rothenberger
- Purdue University College of Pharmacy, Purdue University, West Lafayette, IN
| | - Andra Raibulet
- College of Pharmacy and Health Sciences, Butler University, Indianapolis, IN
| | - James E Slaven
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN
| | - Kristen Suhrie
- Division of Neonatology, Department of Pediatrics, and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Emma M Tillman
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN.
| |
Collapse
|
5
|
Suhrie K, Tillman EM. Pharmacogenetic informed care in early childhood: options for improving access and health equity. Pharmacogenomics 2023; 24:579-582. [PMID: 37466126 DOI: 10.2217/pgs-2023-0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Affiliation(s)
- Kristen Suhrie
- Department of Pediatrics, Division of Neonatology, & Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma M Tillman
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Subasri M, Cressman C, Arje D, Schreyer L, Cooper E, Patel K, Ungar WJ, Barwick M, Denburg A, Hayeems RZ. Translating Precision Health for Pediatrics: A Scoping Review. CHILDREN (BASEL, SWITZERLAND) 2023; 10:897. [PMID: 37238445 PMCID: PMC10217253 DOI: 10.3390/children10050897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023]
Abstract
Precision health aims to personalize treatment and prevention strategies based on individual genetic differences. While it has significantly improved healthcare for specific patient groups, broader translation faces challenges with evidence development, evidence appraisal, and implementation. These challenges are compounded in child health as existing methods fail to incorporate the physiology and socio-biology unique to childhood. This scoping review synthesizes the existing literature on evidence development, appraisal, prioritization, and implementation of precision child health. PubMed, Scopus, Web of Science, and Embase were searched. The included articles were related to pediatrics, precision health, and the translational pathway. Articles were excluded if they were too narrow in scope. In total, 74 articles identified challenges and solutions for putting pediatric precision health interventions into practice. The literature reinforced the unique attributes of children and their implications for study design and identified major themes for the value assessment of precision health interventions for children, including clinical benefit, cost-effectiveness, stakeholder values and preferences, and ethics and equity. Tackling these identified challenges will require developing international data networks and guidelines, re-thinking methods for value assessment, and broadening stakeholder support for the effective implementation of precision health within healthcare organizations. This research was funded by the SickKids Precision Child Health Catalyst Grant.
Collapse
Affiliation(s)
- Mathushan Subasri
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
| | - Celine Cressman
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
| | - Danielle Arje
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
- Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Leighton Schreyer
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
| | - Erin Cooper
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
| | - Komal Patel
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
| | - Wendy J. Ungar
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
- Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
| | - Melanie Barwick
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
- Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
| | - Avram Denburg
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
- Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
- Division of Haematology/Oncology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Robin Z. Hayeems
- Child Health Evaluative Sciences Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; (M.S.); (C.C.); (D.A.); (L.S.); (E.C.); (K.P.); (W.J.U.); (M.B.); (A.D.)
- Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
| |
Collapse
|
7
|
Pharmacogenomics: A Step forward Precision Medicine in Childhood Asthma. Genes (Basel) 2022; 13:genes13040599. [PMID: 35456405 PMCID: PMC9031013 DOI: 10.3390/genes13040599] [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: 02/27/2022] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 02/05/2023] Open
Abstract
Personalized medicine, an approach to care in which individual characteristics are used for targeting interventions and maximizing health outcomes, is rapidly becoming a reality for many diseases. Childhood asthma is a heterogeneous disease and many children have uncontrolled symptoms. Therefore, an individualized approach is needed for improving asthma outcomes in children. The rapidly evolving fields of genomics and pharmacogenomics may provide a way to achieve asthma control and reduce future risks in children with asthma. In particular, pharmacogenomics can provide tools for identifying novel molecular mechanisms and biomarkers to guide treatment. Emergent high-throughput technologies, along with patient pheno-endotypization, will increase our knowledge of several molecular mechanisms involved in asthma pathophysiology and contribute to selecting and stratifying appropriate treatment for each patient.
Collapse
|
8
|
Berrios C, Sadaro SK, Sandritter T, Wagner JA, Soden S, Black B, Abdel-Rahman S. Parental understanding and attitudes following pharmacogenomic testing for pediatric neuropsychiatric patients. Pharmacogenomics 2022; 23:345-354. [DOI: 10.2217/pgs-2022-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study explores parental understanding and attitudes around pharmacogenomic results in their child(ren). Patients and methods: In-depth interviews with parents whose child(ren) had received a pharmacogenomic testing panel for management of neuropsychiatric medications were completed. Interviews were analyzed for themes and accuracy of understanding of results. Results: In 18 parents interviewed, 49/63 (78%) of statements made regarding results were accurate. Differences in understanding were seen by clinic, number of medications and result type. Parents expected results to guide prescribing and perceived the greatest utility in results that could impact current care. Results predicting normal drug metabolism may create mixed feelings. Conclusion: Parents perceive utility in pharmacogenomic testing for their children. Challenges exist in understanding probabilistic and multifactorial information about pharmacogenomic results.
Collapse
Affiliation(s)
- Courtney Berrios
- Genomic Medicine Center, Children's Mercy, Kansas City, MO 64108, USA
- School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Sophia K Sadaro
- Genomic Medicine Center, Children's Mercy, Kansas City, MO 64108, USA
| | - Tracy Sandritter
- Clinical Pharmacology, Toxicology, & Therapeutic Innovation, Children's Mercy, Kansas City, MO 64108, USA
- School of Pharmacy, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Jennifer A Wagner
- School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
- Clinical Pharmacology, Toxicology, & Therapeutic Innovation, Children's Mercy, Kansas City, MO 64108, USA
| | - Sarah Soden
- School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
- Developmental & Behavioral Sciences, Children's Mercy, Kansas City, MO 64108, USA
| | - Benjamin Black
- The Thompson Center for Autism & Neurodevelopmental Disorders, University of Missouri, Columbia, MO 65201, USA
| | - Susan Abdel-Rahman
- Clinical Pharmacology, Toxicology, & Therapeutic Innovation, Children's Mercy, Kansas City, MO 64108, USA
- School of Pharmacy, University of Missouri Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
9
|
Barker CIS, Groeneweg G, Maitland-van der Zee AH, Rieder MJ, Hawcutt DB, Hubbard TJ, Swen JJ, Carleton BC. Pharmacogenomic testing in paediatrics: clinical implementation strategies. Br J Clin Pharmacol 2021; 88:4297-4310. [PMID: 34907575 PMCID: PMC9544158 DOI: 10.1111/bcp.15181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/27/2022] Open
Abstract
Pharmacogenomics (PGx) relates to the study of genetic factors determining variability in drug response. Implementing PGx testing in paediatric patients can enhance drug safety, helping to improve drug efficacy or reduce the risk of toxicity. Despite its clinical relevance, the implementation of PGx testing in paediatric practice to date has been variable and limited. As with most paediatric pharmacological studies, there are well‐recognised barriers to obtaining high‐quality PGx evidence, particularly when patient numbers may be small, and off‐label or unlicensed prescribing remains widespread. Furthermore, trials enrolling small numbers of children can rarely, in isolation, provide sufficient PGx evidence to change clinical practice, so extrapolation from larger PGx studies in adult patients, where scientifically sound, is essential. This review paper discusses the relevance of PGx to paediatrics and considers implementation strategies from a child health perspective. Examples are provided from Canada, the Netherlands and the UK, with consideration of the different healthcare systems and their distinct approaches to implementation, followed by future recommendations based on these cumulative experiences. Improving the evidence base demonstrating the clinical utility and cost‐effectiveness of paediatric PGx testing will be critical to drive implementation forwards. International, interdisciplinary collaborations will enhance paediatric data collation, interpretation and evidence curation, while also supporting dedicated paediatric PGx educational initiatives. PGx consortia and paediatric clinical research networks will continue to play a central role in the streamlined development of effective PGx implementation strategies to help optimise paediatric pharmacotherapy.
Collapse
Affiliation(s)
- Charlotte I S Barker
- Department of Medical & Molecular Genetics, King's College London, London, UK.,Department of Clinical Genetics, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gabriella Groeneweg
- Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Anke H Maitland-van der Zee
- Respiratory Medicine/Pediatric Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael J Rieder
- Departments of Paediatrics, Physiology and Pharmacology and Medicine, Western University, London, Ontario, Canada.,Molecular Medicine Group, Robarts Research Institute, London, Ontario, Canada
| | - Daniel B Hawcutt
- Department of Women's and Children's Health, University of Liverpool, Liverpool, UK.,NIHR Clinical Research Facility, Alder Hey Children's Hospital, Liverpool, UK
| | - Tim J Hubbard
- Department of Medical & Molecular Genetics, King's College London, London, UK.,Genomics England, London, UK
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden, The Netherlands
| | - Bruce C Carleton
- Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Tillman EM, Beavers CJ, Afanasjeva J, Momary KM, Strnad KG, Yerramilli A, Williams AM, Smith BA, Florczykowski B, Fahmy M. Current and future state of clinical pharmacist‐led precision medicine initiatives. JOURNAL OF THE AMERICAN COLLEGE OF CLINICAL PHARMACY 2021. [DOI: 10.1002/jac5.1447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Monica Fahmy
- American College of Clinical Pharmacy Lenexa Kansas USA
| |
Collapse
|
11
|
Qin W, Du Z, Xiao J, Duan H, Shu Q, Li H. Evaluation of clinical impact of pharmacogenomics knowledge involved in CPIC guidelines on Chinese pediatric patients. Pharmacogenomics 2021; 21:209-219. [PMID: 31967514 DOI: 10.2217/pgs-2019-0153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aim: To evaluate the clinical benefits of implementing pharmacogenomics testing for Chinese pediatric patients. Materials & methods : Based on the drug-gene interactions involved in the Clinical Pharmacogenetics Implementation Consortium guidelines, whole-genome sequencing data from the Chinese Academy of Sciences Precision Medicine Initiative project and the medication data of pediatric patients from a children's hospital, the prevalence of the Chinese population with actionable pharmacogenomic variants was calculated, the prescribing pattern for pediatric patients was analyzed. Results: 37.0% of the drugs involved in the Clinical Pharmacogenetics Implementation Consortium guidelines were used by Chinese pediatric patients, 8.91% inpatients and 0.89% outpatients received at least one pharmacogenomics medication, 1.24% (4803) inpatients and 0.16% (2940) outpatients were estimated to be at high risk of pharmacogenomic-related adverse therapeutic outcomes. Conclusion: Implementing pharmacogenomics testing can improve therapeutic outcomes for many Chinese pediatric patients.
Collapse
Affiliation(s)
- Weifeng Qin
- The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou 310052, PR China.,College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Zhenglin Du
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Jingfa Xiao
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Huilong Duan
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Qiang Shu
- The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou 310052, PR China
| | - Haomin Li
- The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou 310052, PR China
| |
Collapse
|
12
|
Turner MA, Cheng K, de Wildt S, Hildebrand H, Attar S, Rossi P, Bonifazi D, Ceci A, Claverol J, Nafria B, Giaquinto C. European research networks to facilitate drug research in children. Br J Clin Pharmacol 2020; 88:4258-4266. [PMID: 32893382 PMCID: PMC9546307 DOI: 10.1111/bcp.14545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/31/2020] [Accepted: 08/16/2020] [Indexed: 01/04/2023] Open
Abstract
Paediatric drug development faces several barriers. These include fragmentation of stakeholders and inconsistent processes during the conduct of research. This review summarises recent efforts to overcome these barriers in Europe. Two exemplar initiatives are described. The European Paediatric Translational Research Infrastructure facilitates preclinical research and other work that underpins clinical trials. conect4children facilitates the design and implementation of clinical trials. Both these initiatives listen to the voices of children and their advocates. Coordination of research needs specific effort that supplements work on science, resources and the policy context.
Collapse
Affiliation(s)
- Mark A Turner
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool Health Partners, Liverpool, UK
| | - Katharine Cheng
- Child Health Innovation Leadership Department (CHILD), Johnson and Johnson, High Wycombe, UK
| | - Saskia de Wildt
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heidrun Hildebrand
- Pediatric Development, Research & Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Sabah Attar
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool Health Partners, Liverpool, UK
| | - Paolo Rossi
- Academic Department of Pediatrics (DPUO), Unit of Perinatal infection and congenital infectious diseases, Bambino Gesu Children's Hospital-University of Rome Tor Vergata, Rome, Italy
| | - Donato Bonifazi
- Consorzio per Valutazioni Biologiche e Farmacologiche, Bari, Italy
| | - Adriana Ceci
- Fondazione per la Ricerca Farmacologica Gianni Benzi Onlus, Bari, Italy
| | - Joana Claverol
- Institut de Recerca Sant Joan de Deu, Sant Joan de Deu Research Foundation, Barcelona, Spain
| | - Begonya Nafria
- Institut de Recerca Sant Joan de Deu, Sant Joan de Deu Research Foundation, Barcelona, Spain
| | - Carlo Giaquinto
- Division of Pediatric Infectious Diseases, Department of Woman's and Child's Health, University of Padua, Padua, Italy
| |
Collapse
|
13
|
Gregornik D, Salyakina D, Brown M, Roiko S, Ramos K. Pediatric pharmacogenomics: challenges and opportunities: on behalf of the Sanford Children's Genomic Medicine Consortium. THE PHARMACOGENOMICS JOURNAL 2020; 21:8-19. [PMID: 32843689 DOI: 10.1038/s41397-020-00181-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 06/15/2020] [Accepted: 08/12/2020] [Indexed: 01/13/2023]
Abstract
The advent of digital, electronic, and molecular technologies has allowed the study of complete genomes. Integrating this information into drug development has opened the door for pharmacogenomic (PGx) interventions in direct patient care. PGx allows clinicians to better identify drug of choice and optimize dosing regimens based on an individual's genetic characteristics. Integrating PGx into pediatric care is a priority for the Sanford Children's Genomic Medicine Consortium, a partnership of ten children's hospitals across the US committed to the innovation and advancement of genomics in pediatric care. In this white paper, we review the current state of PGx research and its clinical utility in pediatrics, a largely understudied population, and make recommendations for advancing cutting-edge practice in pediatrics.
Collapse
Affiliation(s)
- David Gregornik
- Pharmacogenomics Program, Children's Minnesota, Minneapolis, MN, USA.
| | - Daria Salyakina
- Research Institute and Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL, USA
| | - Marilyn Brown
- Research Institute and Personalized Medicine Initiative, Nicklaus Children's Hospital, Miami, FL, USA
| | - Samuel Roiko
- Children's Research Institute, Minnesota, Minneapolis, MN, USA
| | - Kenneth Ramos
- Texas A&M University System, College Station, TX, USA.
| |
Collapse
|
14
|
Onland W, Hutten J, Miedema M, Bos LD, Brinkman P, Maitland-van der Zee AH, van Kaam AH. Precision Medicine in Neonates: Future Perspectives for the Lung. Front Pediatr 2020; 8:586061. [PMID: 33251166 PMCID: PMC7673376 DOI: 10.3389/fped.2020.586061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of pre-term birth with long lasting sequelae. Since its first description more than 50 years ago, many large randomized controlled trials have been conducted, aiming to improve evidence-based knowledge on the optimal strategies to prevent and treat BPD. However, most of these intervention studies have been performed on a population level without regard for the variation in clinical and biological diversity (e.g., gestational age, ethnicity, gender, or disease progression) between patients that is driven by the complex interaction of genetic pre-disposition and environmental exposures. Nevertheless, clinicians provide daily care such as lung protective interventions on an individual basis every day despite the fact that research supporting individualized or precision medicine for monitoring or treating pre-term lungs is immature. This narrative review summarizes four potential developments in pulmonary research that might facilitate the process of individualizing lung protective interventions to prevent development of BPD. Electrical impedance tomography and electromyography of the diaphragm are bedside monitoring tools to assess regional changes in lung volume and ventilation and spontaneous breathing effort, respectively. These non-invasive tools allow a more individualized optimization of invasive and non-invasive respiratory support. Investigation of the genomic variation in caffeine metabolism in pre-term infants can be used to optimize and individualize caffeine dosing regimens. Finally, volatile organic compound analysis in exhaled breath might accurately predict BPD at an early stage of the disease, enabling clinicians to initiate preventive strategies for BPD on an individual basis. Before these suggested diagnostic or monitoring tools can be implemented in daily practice and improve individualized patient care, future research should address and overcome their technical difficulties, perform extensive external validation and show their additional value in preventing BPD.
Collapse
Affiliation(s)
- Wes Onland
- Department of Neonatology, Amsterdam University Medical Centers, VU University Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Jeroen Hutten
- Department of Neonatology, Amsterdam University Medical Centers, VU University Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Martijn Miedema
- Department of Neonatology, Amsterdam University Medical Centers, VU University Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Lieuwe D Bos
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Paul Brinkman
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Anton H van Kaam
- Department of Neonatology, Amsterdam University Medical Centers, VU University Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
15
|
Gogou M, Pavlou E. Efficacy of antiepileptic drugs in the era of pharmacogenomics: A focus on childhood. Eur J Paediatr Neurol 2019; 23:674-684. [PMID: 31280948 DOI: 10.1016/j.ejpn.2019.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/23/2019] [Accepted: 06/24/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND In recent years advances in the field of pharmacogenomics have expanded the concept for more individualized treatments. Our aim is to provide literature data about the relationship between genetic polymorphisms and efficacy of antiepileptic drugs in children. METHODS Pubmed was used as the main medical database source. Only original research papers were considered. No year-of-publication restriction was placed. Quality of evidence was assessed according to American Academy of Neurology guidelines. RESULTS A total of 12 cross-sectional and case-control studies fulfilled our selection criteria. ABCB1 gene was associated with drug responsiveness in 2 out of 6 studies and ABCC2 gene in 1 out of 1 studies. SCN1A gene was also associated with seizure control in 4 out of 5 studies. Cytochrome P450 genes were found to significantly affect drug responsiveness in 2 out of 4 studies, while polymorphisms of uridinediphosphateglucuronosyltransferaseUGT2B7 gene predisposed to drug-resistance in 1 out of 2 studies. CONCLUSION Variability in genes coding for sodium channels, drug transporters and cytochrome P450 enzymes can have a significant impact on response to antiepileptic drugs. Larger prospective studies with better stratification of samples are needed to shed light on these associations.
Collapse
Affiliation(s)
- Maria Gogou
- 2nd Department of Pediatrics, University General Hospital AHEPA, Thessaloniki, Greece.
| | - Evangelos Pavlou
- 2nd Department of Pediatrics, University General Hospital AHEPA, Thessaloniki, Greece
| |
Collapse
|
16
|
Ta R, Cayabyab MA, Coloso R. Precision medicine: a call for increased pharmacogenomic education. Per Med 2019; 16:233-245. [PMID: 31025601 DOI: 10.2217/pme-2018-0107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Precision medicine is an emerging model of care where providers consider patients' genetic profiles, lifestyles and environments to offer more precise therapy. The potential of precision medicine is boundless as interdisciplinary teams utilize genetic technologies to improve patient outcomes. The integration of precision medicine into healthcare faces many barriers, including a lack of standardization and reimbursement concerns. This article argues that increased pharmacogenetics education and system-wide implementation is necessary to overcome some of these challenges. Extensive expansion of pharmacogenomics education is a step toward producing knowledgeable clinicians who are poised to apply its methodology and champion for patient-centered care.
Collapse
Affiliation(s)
- Richard Ta
- University of California, San Francisco, School of Pharmacy, Class of 2020; San Francisco, CA, 94143, USA
| | - Mari As Cayabyab
- University of California, San Francisco, School of Pharmacy, Class of 2020; San Francisco, CA, 94143, USA
| | - Rodolfo Coloso
- University of California, San Francisco, School of Pharmacy, Class of 2021P; San Francisco, CA, 94143, USA
| |
Collapse
|
17
|
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
|
18
|
Wright GEB, Amstutz U, Drögemöller BI, Shih J, Rassekh SR, Hayden MR, Carleton BC, Ross CJD. Pharmacogenomics of Vincristine-Induced Peripheral Neuropathy Implicates Pharmacokinetic and Inherited Neuropathy Genes. Clin Pharmacol Ther 2018; 105:402-410. [PMID: 29999516 PMCID: PMC6519044 DOI: 10.1002/cpt.1179] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022]
Abstract
Vincristine is an effective chemotherapeutic drug for various cancers, including acute lymphoblastic leukemia (ALL). Unfortunately, clinical utility is restricted by dose‐limiting vincristine‐induced peripheral neuropathies (VIPN). We sought to determine the association of VIPN with a recently identified risk variant, CEP72 rs924607, and drug absorption, distribution, metabolism, and excretion (ADME) gene variants in pediatric ALL. This was followed by a meta‐analysis of pharmacogenomic data from over 500 patients. CEP72 rs924607 was significantly associated with VIPN (P = 0.02; odds ratio (OR) = 3.4). ADME analyses identified associations between VIPN and ABCC1 rs3784867 (P = 5.34 × 10−5; OR = 4.9), and SLC5A7 rs1013940 (P = 9.00 × 10−4; OR= 8.6); genes involved in vincristine transport and inherited neuropathies, respectively. Meta‐analysis identified an association with a variant related to TTPA (rs10504361: P = 6.85 × 10−4; OR = 2.0), a heritable neuropathy‐related gene. This study provides essential corroboratory evidence for CEP72 rs924607 and highlights the importance of drug transporter and inherited neuropathy genes in VIPN.
Collapse
Affiliation(s)
- Galen E B Wright
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Ursula Amstutz
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,University Institute of Clinical Chemistry, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Britt I Drögemöller
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanne Shih
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Shahrad R Rassekh
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael R Hayden
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
19
|
Adam de Beaumais T, Jacqz-Aigrain E. Pharmacogenetics: Applications to Pediatric Patients. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 83:191-215. [PMID: 29801575 DOI: 10.1016/bs.apha.2018.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Individual genomic differences may affect drug disposition and effects of many drugs, and identification of biomarkers are crucial to personalize dosage and optimize response. In children, developmental changes associated with growth and maturation translate into different relationships between genotype and phenotype and different responses to treatment compared to adults. This review aims to summarize some developmental aspects of pharmacogenetics, based on practical examples.
Collapse
Affiliation(s)
- Tiphaine Adam de Beaumais
- Department of Paediatric Pharmacology and Pharmacogenetics, Robert Debré Hospital, APHP, Paris, France
| | - Evelyne Jacqz-Aigrain
- Department of Paediatric Pharmacology and Pharmacogenetics, Robert Debré Hospital, APHP, Paris, France; University Paris Diderot Sorbonne Paris Cité, Paris, France; Clinical Investigation Center CIC1426, INSERM, Paris, France.
| |
Collapse
|
20
|
Zhou H, Li L, Yang P, Yang L, Zheng JE, Zhou Y, Han Y. Optimal predictor for 6-mercaptopurine intolerance in Chinese children with acute lymphoblastic leukemia: NUDT15, TPMT, or ITPA genetic variants? BMC Cancer 2018; 18:516. [PMID: 29720126 PMCID: PMC5932771 DOI: 10.1186/s12885-018-4398-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/18/2018] [Indexed: 12/16/2022] Open
Abstract
Background 6-mercaptopurine (6-MP) contributes substantially to remarkable improvement in the survival of childhood acute lymphoblastic leukemia (ALL) patients. However, 6-MP also has dose-limiting toxicities, particularly life-threatening myelosuppression, due to genetic polymorphisms in enzymes that metabolize 6-MP. Promising biomarkers for predicting 6-MP-induced leukopenia is still unclear in Chinese population. Here, we evaluated the associations of NUDT15, TPMT and ITPA genotypes with 6-MP intolerance in our cohort of childhood ALL patients. Methods A total of 105 Chinese pediatric patients with a confirmed diagnosis of ALL were enrolled. We identified the NUDT15 coding variant rs116855232 (c.415C > T), a newly discovered 6-MP toxicity-related locus in Asians, and polymorphisms in TPMT rs1142345 and ITPA rs11273540. Associations between genotypes and 6-MP dose sensitivity, leukopenia, hepatotoxicity, and therapy interruption were evaluated. Results The minor allele frequencies (MAFs) of NUDT15 rs116855232, TPMT rs1142345 and ITPA rs11273540 were 15.7, 2.9, and 18.1%, respectively. NUDT15 and TPMT genetic variants were strongly associated with 6-MP dose intensity. Patients with NUDT15 homogenous genotype (TT) were highly sensitive to 6-MP (dose intensity of 60.27%) compared to these with heterozygous genotype (TC) or wild type (CC), who tolerated an average dose intensity of 83.83 and 94.24%, respectively. The NUDT15 variant was a predictor for leukopenia (OR: 3.62, 95% CI 1.377–9.501, P = 0.009) and early-onset leukopenia (OR: 9.63, 95% CI 2.764–33.514, P = 3.75 × 10− 4). No differences were found between 6-MP dose intensity and ITPA polymorphisms. Conclusion NUDT15 variant is an optimal predictor for 6-MP intolerance in Chinese pediatric ALL patients and may have greatly clinical implications for individualized therapy.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong, University of Science and Technology, No. 1277, Jie Fang Road, Wuhan, 430022, China
| | - Lei Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, 430022, China
| | - Peng Yang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Yang
- Department of Pharmacy, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Jin E Zheng
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ying Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong, University of Science and Technology, No. 1277, Jie Fang Road, Wuhan, 430022, China
| | - Yong Han
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong, University of Science and Technology, No. 1277, Jie Fang Road, Wuhan, 430022, China.
| |
Collapse
|
21
|
Slob EMA, Vijverberg SJH, Pijnenburg MW, Koppelman GH, Maitland-van der Zee AH. What do we need to transfer pharmacogenetics findings into the clinic? Pharmacogenomics 2018; 19:589-592. [PMID: 29701121 DOI: 10.2217/pgs-2018-0026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Elise M A Slob
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, P.O. Box 22700, NL-1100 DE Amsterdam, The Netherlands
| | - Susanne J H Vijverberg
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, P.O. Box 22700, NL-1100 DE 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, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Groningen, The Netherlands.,Groningen Research Institute for Asthma & COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anke-Hilse Maitland-van der Zee
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, P.O. Box 22700, NL-1100 DE Amsterdam, The Netherlands
| |
Collapse
|
22
|
Leroux S, Elie V, Zhao W, Magreault S, Jacqz-Aigrain E. Principles and applications of pharmacometrics in drug evaluation in children. Therapie 2018; 73:165-170. [DOI: 10.1016/j.therap.2017.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022]
|
23
|
Abstract
Identification of markers involved in drug disposition is crucial for drugs with a narrow therapeutic index. Individual genomic differences can affect the pharmacology of some drugs and participate to inter-individual variability in drug response. Pharmacogenetics is a useful tool in clinical practice for dosage adjustment and to limit drug toxicities. In pediatrics, physiological changes can also influence the disposition of drugs in infants, children and adolescents. The importance of ontogeny translates into different responses to the same drug in children and adults. Thus, interactions between the maturation of metabolism enzymes or transporters and genetics have a major impact on drug exposure leading to age-specific dosage requirements. This review aims to describe implementation of pharmacogenetics in personalized medicine and specifies pediatric characteristics with ethical considerations.
Collapse
Affiliation(s)
- Virginia Neyro
- Department of paediatric pharmacology and pharmacogenetics, Robert-Debré hospital, AP-HP, 75019 Paris, France
| | - Evelyne Jacqz-Aigrain
- Department of paediatric pharmacology and pharmacogenetics, Robert-Debré hospital, AP-HP, 75019 Paris, France; University of Paris Diderot Sorbonne Paris Cité, 75013 Paris, France; Clinical investigation center (CIC1426), Inserm, 75019 Paris, France
| | - Tiphaine Adam de Beaumais
- Department of paediatric pharmacology and pharmacogenetics, Robert-Debré hospital, AP-HP, 75019 Paris, France; Precision cancer medicine team, Gustave-Roussy, 94800 Villejuif, France.
| |
Collapse
|
24
|
Lui JK, Lutchen KR. The role of heterogeneity in asthma: a structure-to-function perspective. Clin Transl Med 2017; 6:29. [PMID: 28776171 PMCID: PMC5543015 DOI: 10.1186/s40169-017-0159-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
A number of methods have evolved through the years in probing the dysfunction that impacts mechanics and ventilation in asthma. What has been consistently found is the notion of heterogeneity that is not only captured in the frequency dependence of lung mechanics measurements but also rendered on imaging as patchy diffuse areas of ventilation defects. The degree of heterogeneity has been linked to airway hyperresponsiveness, a hallmark feature of asthma. How these heterogeneous constriction patterns lead to functional impairment in asthma have only been recently explored using computational airway tree models. By synthesizing measurements of lung mechanics and advances in imaging, computational airway tree models serve as a powerful engine to accelerate our understanding of the physiologic changes that occur in asthma. This review will be focused on the current state of investigational work on the role of heterogeneity in asthma, specifically exploring the structural and functional relationships.
Collapse
Affiliation(s)
- Justin K. Lui
- Department of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Kenneth R. Lutchen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215 USA
| |
Collapse
|
25
|
Matera MG, Rinaldi B, Calzetta L, Cazzola M. Pharmacogenetic and pharmacogenomic considerations of asthma treatment. Expert Opin Drug Metab Toxicol 2017; 13:1159-1167. [PMID: 28992739 DOI: 10.1080/17425255.2017.1391215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Pharmacogenetic and pharmacogenomic approaches are already utilized in some areas, such as oncology and cardiovascular disease, for selecting appropriate patients and/or establishing treatment and dosing guidelines. This is not true in asthma although many patients have different responses to drug treatment due to genetic factors. Areas covered: Several genetic factors that affect the pharmacotherapeutic responses to asthma medications, such as β2-AR agonists, corticosteroids, and leukotriene modifiers and could contribute to significant between-person variability in response are described. Expert opinion: An expanding number of genetic loci have been associated with therapeutic responses to asthma drugs but the individual effect of one single-nucleotide polymorphism is partial. In fact, epigenetic changes can modify genetic effects in time-, environment-, and tissue-specific manners, genes interact together in networks, and nongenetic components such as environmental exposures, gender, nutrients, and lifestyle can significantly interact with genetics to determine the response to therapy. Therefore, well-designed randomized controlled trials or observational studies are now mandatory to define if response to asthma medications in individual patients can be improved by using pharmacogenetic predictors of treatment response. Meanwhile, routine implementation of pharmacogenetics and pharmacogenomics into clinical practice remains a futuristic, far-off challenge for many clinical practices.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- a Department of Experimental Medicine , University of Campania Luigi Vanvitelli , Naples , Italy
| | - Barbara Rinaldi
- a Department of Experimental Medicine , University of Campania Luigi Vanvitelli , Naples , Italy
| | - Luigino Calzetta
- b Department of Systems Medicine , University of Rome Tor Vergata , Rome , Italy
| | - Mario Cazzola
- b Department of Systems Medicine , University of Rome Tor Vergata , Rome , Italy
| |
Collapse
|
26
|
Mennella JA, Mathew PS, Lowenthal ED. Use of Adult Sensory Panel to Study Individual Differences in the Palatability of a Pediatric HIV Treatment Drug. Clin Ther 2017; 39:2038-2048. [PMID: 28923290 PMCID: PMC5654675 DOI: 10.1016/j.clinthera.2017.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/26/2017] [Accepted: 08/23/2017] [Indexed: 01/17/2023]
Abstract
PURPOSE The recommended first-line treatment for young children infected with HIV includes the liquid formulation of the co-formulated protease inhibitors lopinavir/ritonavir (Kaletra® [Abbott Laboratories, Chicago, Illinois]). Clinical reports indicate that some children readily accept the taste of Kaletra, whereas others strongly reject it, which can deter therapeutic adherence and outcomes. METHODS As a proof-of-concept approach, a sensory panel of genotyped adults was used to document the range of individual differences in the taste and palatability (hedonics) of the liquid formulation of Kaletra and other taste stimuli, including common excipients. Panelists rated taste sensations using generalized labeled magnitude scales to determine genotype-phenotype relationships. Several months later, the panelists were retested to assess response reliability. FINDINGS Not all panelists had the same sensory experience when tasting Kaletra. Palatability ratings varied widely, from moderate like to strongest imaginable dislike, and were reliable over time. The more irritating and bitter Kaletra tasted, the more disliked by the panelist. The more they disliked the taste of Kaletra, the more they disliked the taste of its excipient ethanol and the bitter stimulus denatonium. Those who experienced less bitter and sweeter taste sensations had a different genetic signature than the other panelists. Bitterness and irritation ratings of Kaletra varied by the orphaned bitter receptor gene (TAS2R60), whereas sweetness ratings of Kaletra varied according to the cold receptor gene (TRPM8), which is activated by menthol, an excipient of Kaletra. Neither genotype related to ratings for ethanol or denatonium, however. IMPLICATIONS The use of a sensory panel holds promise as a first step in determining the nature of individual differences in the palatability of existing pediatric drug formulations and sources of variation. In this era of personalized medicine, the need is great to develop psychophysical tools to determine which drugs will show variation in acceptance by children and whether patterns of individual variation in taste as assessed by adults mirror those of young patients. ClinicalTrials.gov identifier: NCT01841710.
Collapse
Affiliation(s)
| | | | - Elizabeth D Lowenthal
- University of Pennsylvania Perelman School of Medicine, Departments of Pediatrics and Epidemiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
L-Methylfolate supplementation in a child with autism and methyltetrahydrofolate reductase, enzyme gene C677TT allele. Psychiatr Genet 2017; 27:116-119. [DOI: 10.1097/ypg.0000000000000170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Al-Mahayri ZN, Patrinos GP, Ali BR. Pharmacogenomics in pediatric acute lymphoblastic leukemia: promises and limitations. Pharmacogenomics 2017; 18:687-699. [PMID: 28468529 DOI: 10.2217/pgs-2017-0005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite the significant advances achieved in pediatric acute lymphocytic leukemia (ALL) treatment, adverse side effects of drugs remain a challenging issue. Numerous ALL pharmacogenomic studies have been conducted to elucidate the predisposing genetic factors for their development. Plausible pharmacogenomic data are available for the osteonecrosis associated with glucocorticoids, the neurotoxicity associated with vincristine and the cardiotoxicity related to anthracyclines. However, these data have not been fully translated into the clinic due to several limitations, most importantly the lack of reliable evidence. The most robust pharmacogenomics data are those for thiopurines and methotrexate use, with evidence-based preemptive testing recommendations for the former. Pharmacogenomics has a significant potential utility in pediatric ALL treatment regimens. In this review, gaps and limitations in this field are emphasized, which may provide a useful guide for future research design.
Collapse
Affiliation(s)
- Zeina N Al-Mahayri
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, United Arab Emirates
| | - George P Patrinos
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, United Arab Emirates.,Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, Patras, Greece
| | - Bassam R Ali
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, United Arab Emirates
| |
Collapse
|
29
|
Elzagallaai AA, Greff M, Rieder MJ. Adverse Drug Reactions in Children: The Double-Edged Sword of Therapeutics. Clin Pharmacol Ther 2017; 101:725-735. [PMID: 28295234 DOI: 10.1002/cpt.677] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/14/2022]
Abstract
Adverse drug reactions (ADRs) represent a major health problem worldwide, with high morbidity and mortality rates. ADRs are classified into Type A (augmented) and Type B (bizarre) ADRs, with the former group being more common and the latter less common but often severe and clinically more problematic due to their unpredictable nature and occurrence at any dose. Pediatric populations are especially vulnerable to ADRs due to the lack of data for this age group from the drug development process and because of the wide use of off-label and unlicensed use of drugs. Children are more prone to specific types of ADRs because of the level of maturity of body systems involved in absorption, metabolism, transportation, and elimination of drugs. This state-of-the-art review provides an overview of definitions, classifications, epidemiology, and pathophysiology of ADRs and discusses the available evidence for related risk factors and causes of ADRs in the pediatric population.
Collapse
Affiliation(s)
- A A Elzagallaai
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada.,Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Mje Greff
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada.,Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - M J Rieder
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada.,Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Ontario, Canada
| |
Collapse
|
30
|
van der Wouden CH, Cambon-Thomsen A, Cecchin E, Cheung KC, Dávila-Fajardo CL, Deneer VH, Dolžan V, Ingelman-Sundberg M, Jönsson S, Karlsson MO, Kriek M, Mitropoulou C, Patrinos GP, Pirmohamed M, Samwald M, Schaeffeler E, Schwab M, Steinberger D, Stingl J, Sunder-Plassmann G, Toffoli G, Turner RM, van Rhenen MH, Swen JJ, Guchelaar HJ. Implementing Pharmacogenomics in Europe: Design and Implementation Strategy of the Ubiquitous Pharmacogenomics Consortium. Clin Pharmacol Ther 2017; 101:341-358. [DOI: 10.1002/cpt.602] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022]
Affiliation(s)
- CH van der Wouden
- Department of Clinical Pharmacy and Toxicology; Leiden University Medical Center; Leiden The Netherlands
| | - A Cambon-Thomsen
- UMR Inserm U1027 and Université de Toulouse III Paul Sabatier; Toulouse France
| | - E Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico; National Cancer Institute; Aviano Italy
| | - KC Cheung
- Royal Dutch Pharmacists Association (KNMP); The Hague The Netherlands
| | - CL Dávila-Fajardo
- Department of Clinical Pharmacy, Granada University Hospital; Institute for Biomedical Research; Granada Spain
| | - VH Deneer
- Department of Clinical Pharmacy; St Antonius Hospital; Nieuwegein The Netherlands
| | - V Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine; University of Ljubljana; Slovenia
| | - M Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics; Karolinska Institutet; Stockholm Sweden
| | - S Jönsson
- Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| | - MO Karlsson
- Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| | - M Kriek
- Center for Clinical Genetics; Leiden University Medical Center; Leiden The Netherlands
| | | | - GP Patrinos
- University of Patras, School of Health Sciences, Department of Pharmacy; University Campus; Rion Patras Greece
| | - M Pirmohamed
- Department of Molecular and Clinical Pharmacology; Royal Liverpool University Hospital and University of Liverpool; Liverpool United Kingdom
| | - M Samwald
- Center for Medical Statistics, Informatics, and Intelligent Systems; Medical University of Vienna; Vienna Austria
| | - E Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart; Germany and University of Tübingen; Tübingen Germany
| | - M Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart; Germany and University of Tübingen; Tübingen Germany
- Department of Clinical Pharmacology; University Hospital Tübingen; Tübingen Germany
- Department of Pharmacy and Biochemistry; University of Tübingen; Tübingen Germany
| | - D Steinberger
- Bio.logis Center for Human Genetics; Frankfurt am Main Germany
| | - J Stingl
- Research Division; Federal Institute for Drugs and Medical Devices; Bonn Germany
| | - G Sunder-Plassmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III; Medical University of Vienna; Vienna Austria
| | - G Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico; National Cancer Institute; Aviano Italy
| | - RM Turner
- Department of Molecular and Clinical Pharmacology; Royal Liverpool University Hospital and University of Liverpool; Liverpool United Kingdom
| | - MH van Rhenen
- Royal Dutch Pharmacists Association (KNMP); The Hague The Netherlands
| | - JJ Swen
- Department of Clinical Pharmacy and Toxicology; Leiden University Medical Center; Leiden The Netherlands
| | - H-J Guchelaar
- Department of Clinical Pharmacy and Toxicology; Leiden University Medical Center; Leiden The Netherlands
| | | |
Collapse
|