Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-27T10:11:27.965Z Has data issue: false hasContentIssue false

22 - Application of Pharmacogenetics and Pharmacogenomics in Pediatrics: What Makes Children Different?

from II - Therapeutic Areas

Published online by Cambridge University Press:  05 June 2012

Russ B. Altman
Affiliation:
Stanford University, California
David Flockhart
Affiliation:
Indiana University
David B. Goldstein
Affiliation:
Duke University, North Carolina
Get access

Summary

Historically, submissions to the Food and Drug Administration (FDA) have been based on safety and efficacy data obtained from clinical trials conducted in adults with limited or no data from children. As a result, pediatricians and other health care professionals have relied on empiric therapeutic strategies, largely the consequence of treatment on a trial-and-error basis. In essence, the absence of information in the product label forces pediatricians to choose between avoiding the use of a medication that may be beneficial and using a medication “off-label” in the absence of evidence-based safety and efficacy data with the accompanying potential for ineffective and harmful outcomes.

During the past fifteen years, new federal laws and regulations have increased the level of scientific and clinical rigor of investigations aimed at ensuring that the use of medications by children is, indeed, safe and effective. Interested readers are referred to a detailed chronology of events occurring between 1994 and 2002 (1), and a contemporary discussion of the issues surrounding more recent legislative activities, such as the Pediatric Research Equity Act (PREA) of January 2003 (2). In general, it is recognized that growth and development are accompanied by changes in the physiological and biochemical processes determining drug disposition and response, for example, drug absorption, distribution, metabolism, excretion, and targets of drug response (3). However, the acquisition of information that can inform safe and efficacious use of medications in children of different ages or developmental stages has been a relatively recent development, increasing dramatically in recent years as a consequence of the various legislative initiatives. It is now apparent that extrapolation of adult data to pediatric populations is quite inappropriate because drug clearance may be greater than or, in some cases, less than that observed in adults (4). Thus, even though weight-based dosing strategies are becoming more sophisticated and have improved abilities to use adult data to infer drug clearance in children (5), they are unlikely to provide consistent dosing guidelines across all pediatric age groups or chemical classes. This is largely due to the variability in the developmental patterns of expression of the various drug-metabolizing enzymes and transporters involved in the disposition of individual compounds. Furthermore, evidence that the response to some medications may be different in children relative to adults despite comparable drug exposure is beginning to accrue (e.g., buspirone [4]), implying that age-related differences in drug targets and downstream signal transduction pathways may also be present.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2012

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Steinbrook, RTesting medications in childrenN Engl J Med 2002 347 1462Google Scholar
Ward, RMKaufman, RFuture of pediatric therapeutics: reauthorization of BPCA and PREAClin Pharmacol Ther 2008 81 477Google Scholar
Kearns, GLAbdel-Rahman, SMAlander, SWBlowey, DLLeeder, JSKauffman, REThe impact of ontogeny on drug disposition and actionN Engl J Med 2003 349 1157Google Scholar
Rodriguez, WSelen, AAvant, DImproving pediatric dosing through pediatric initiatives: what have we learnedPediatrics 2008 121 530Google Scholar
Anderson, BJHolford, NHGMechanism-based concepts of size and maturity in pharmacokineticsAnnu Rev Pharmacol Toxicol 2008 48 303Google Scholar
Weinshilboum, RMWang, LPharmacogenetics and pharmacogenomics: development, science and translationAnnu Rev Genomics Hum Genet 2006 7 223Google Scholar
Leeder, JSDevelopmental and pediatric pharmacogenomicsPharmacogenomics 2003 4 331Google Scholar
Hines, RNThe ontogeny of drug metabolism enzymes and implications for adverse drug eventsPharmacol Ther 2008 118 250Google Scholar
Cheok, MEvans, WEAcute lymphoblastic leukaemia: a model for the pharmacogenomics of cancer therapyNat Rev Cancer 2006 6 117Google Scholar
Husain, ALoehle, JAHein, DWClinical pharmacogenetics in pediatric patientsPharmacogenomics 2007 8 1403Google Scholar
Kronenberg, SFrisch, ARotberg, BCarmel, MApter, AWeizman, APharmacogenetics of selective serotonin reuptake inhibitors in pediatric depression and anxietyPharmacogenomics 2008 9 1725Google Scholar
Nebert, DWRussell, DWClinical importance of the cytochromes P450Lancet 2002 360 1155Google Scholar
Coughtrie, MWHSulfation through the looking glass – recent advances in sulfotransferase research for the curiousPharmacogenomics J 2002 2 297Google Scholar
Choudhuri, SKlaassen, CDStructure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), ABCG2 (BCRP) efflux transportersInt J Toxicol 2006 25 231Google Scholar
Hagenbuch, BGui, CXenobiotic transporters of the human organic anion transporting polypeptides (OATP) familyXenobiotica 2008 38 778Google Scholar
Williams, JARing, BJCantrell, VEComparative metabolic capabilities of CYP3A4, CYP3A5 and CYP3A7Drug Metab Disp 2002 30 883Google Scholar
Witorsch, RJLow-dose in utero effects of xenoestrogens in mice and their relevance to humans: as analytical review of the literatureFood Chem Toxicol 2002 40 905Google Scholar
Kitada, MKamataki, TItahashi, KRikihisa, TKanakubo, YP-450 HFLa, a form of cytochrome P-450 purified from human fetal livers, is the 16α-hydroxylase of dehydroepiandrosterone 3-sulfateJ Biol Chem 1987 262 13534Google Scholar
Lin, ZLFontaine, JWatchko, JFCoexpression of gene polymorphisms involved in bilirubin production and metabolismPediatrics 2008 122 e156Google Scholar
French, DWang, WCheng, CAcquired variation outweighs inherited variation in whole genome analysis of methotrexate polyglutamate accumulation in leukemiaBlood 2009Google Scholar
Alcorn, JMcNamara, PJOntogeny of hepatic and renal systemic clearance pathways in infantsClin Pharmacokinet 2002 41 959Google Scholar
Alcorn, JMcNamara, PJOntogeny of hepatic and renal systemic clearance pathways in infantsClin Pharmacokinet 2002 41 1077Google Scholar
Ring, JAGhabrial, HChing, MSSmallwood, RAMorgan, DJFetal hepatic drug eliminationPharmacol Ther 1999 84 429Google Scholar
Koukouritaki, SBManro, JRMarsh, SADevelopmental expression of human hepatic CYP2C9 and CYP2C19J Pharmacol Exp Ther 2004 308 965Google Scholar
Blake, MJGaedigk, APearce, REOntogeny of dextromethorphan O- and N-demethylation in the first year of lifeClin Pharmacol Ther 2007 81 510Google Scholar
WagnerWeiner, LPediatric rheumatology for the adult rheumatologistJ Clin Rheumatol 2008 14 109Google Scholar
Gelfand, EWIs asthma in childhood different from asthma in adults? Why do we need special approaches to asthma in children?Allergy Asthma Proc 2008 29 99Google Scholar
Flynn, JTMitsnefes, MPierce, CBlood pressure in children with chronic kidney disease. A report from the Chronic Kidney Disease in Children StudyHypertension 2008 52 1Google Scholar
Hinks, ABarton, AShephard, NIdentification of a novel susceptibility locus for juvenile idiopathic arthritis by genome-wide association analysisArthritis Rheum 2009 60 258Google Scholar
Kugathasan, SBaldassano, RNBradfield, JPLoci on 20q13 and 21q22 are associated with pediatric-onset inflammatory bowel diseaseNat Genet 2008 40 1211Google Scholar
Moffatt, MFKabesch, MLiang, LGenetic variants regulating ORMDL3 expression contribute to the risk of childhood asthmaNature 2007 448 470Google Scholar
Burgner, DDavila, SBreunis, WBA genome-wide association study identifies novel and functionally related susceptibility loci for Kawasaki diseasePLoS Genet 2009 5Google Scholar
Bouzigon, ECorda, EAschard, HEffect of 17q21 variants and smoking exposure in early-onset asthmaN Engl J Med 2008 359 1985Google Scholar
Guberman, AHBesag, FMCBrodie, MJLamotrigine-induced rash: risk/benefit considerations in adults and childrenEpilepsia 1999 40 985Google Scholar
Relling, MVBoyett, JMBlanco, JGGranulocyte colony-stimulating factor and the risk of secondary myeloid malignancy after etoposide treatmentBlood 2003 101 3862Google Scholar
Edick, MJCheng, CYang, WLymphoid gene expression as a predictor of risk of secondary brain tumorsGenes Chromosomes Cancer 2005 42 107Google Scholar
Hartford, CYang, WCheng, CGenome scan implicates adhesion biological pathways in secondary leukemiaLeukemia 2007 21 2128Google Scholar
Food and Drug Modernization Act of 1997 1997
Best Pharmaceuticals for Children Act 2002
Pediatric Research Equity Act of 2003 2003
AbdelRahman, SMReed, MDWells, TGKearns, GLConsiderations in the rational design and conduct of phase I/II pediatric clinical trials: avoiding the problems and pitfallsClin Pharmacol Ther 2007 81 483Google Scholar
Benjamin, DKJSmith, PBJadhav, PPediatric antihypertensive trial failures: analysis of end points and dose rangeHypertension 2008 51 834Google Scholar
Chládek, JZimová, GMartínková, JTuma, IIntra-individual variability and influence of urine collection period on dextromethorphan metabolic ratios in healthy subjectsFundam Clin Pharmacol 1999 13 508Google Scholar
Jones, DRGorski, JCHamman, MAHall, SDQuantification of dextromethorphan and metabolites: a dual phenotypic marker for cytochrome P450 3A4/5 and 2D6 activityJ Chromatogr B 1996 678 105Google Scholar
Kerry, NLSomogyi, AABochner, FMikus, GThe role of CYP2D6 in primary and secondary oxidative metabolism of dextromethorphan: studies using human liver microsomesBr J Clin Pharmacol 1994 38 243Google Scholar
Von Moltke, LLGreenblatt, DJGrassi, JMMultiple human cytochromes contribute to biotransformation of dextromethorphan in-vitro: role of CYP2C9, CYP2C19, CYP2D6, and CYP3AJ Pharm Pharmacol 1998 50 997Google Scholar
Wang, YUnadkat, JDEnzymes in addition to CYP3A4 and 3A5 mediate -demethylation of dextromethorphan in human liver microsomesBiopharm Drug Dispos 1999 20 341Google Scholar
Capon, DABochner, FKerry, NMikus, GDanz, CSomogyi, AAThe influence of CYP2D6 polymorphism and quinidine on the disposition and antitussive effect of dextromethorphan in humansClin Pharmacol Ther 1996 60 295Google Scholar
Pope, LEKhalil, MHBerg, JEStiles, MYakatan, GJSellers, EMPharmacokinetics of dextromethorphan after single or multiple dosing in combination with quinidine in extensive and poor metabolizersJ Clin Pharmacol 2004 44 1132Google Scholar
Kanaan, MDaali, YDayer, PDesmeules, JLack of interaction of the NMDA receptor antagonists dextromethorphan and dextrorphan with P-glycoproteinCurr Drug Metab 2008 9 144Google Scholar
Frank, DJaehde, UFuhr, UEvaluation of probe drugs and pharmacokinetic metrics for CYP2D6 phenotypingEur J Clin Pharmacol 2007 63 321Google Scholar
Treluyer, J-MJacqz-Aigrain, EAlvarez, FCresteil, TExpression of in developing human liverEur J Biochem 1991 202 583Google Scholar
Stevens, JCMarsh, SAZaya, MJDevelopmental changes in human liver CYP2D6 expressionDrug Metab Dispos 2008 36 1587Google Scholar
Relling, MVCherrie, JSchell, MJPetros, WPMeyer, WHEvans, WELower prevalence of the debrisoquin oxidative poor metabolizer phenotype in American black versus white subjectsClin Pharmacol Ther 1991 50 308Google Scholar
Zanger, UMRaimundo, SEichelbaum, MCytochrome P450 2D6: overview and update on pharmacology, genetics and biochemistryNaunyn-Schmiedeberg's Arch Pharmacol 2004 369 23Google Scholar
IngelmanSundberg, MGenetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversityPharmacogenomics J 2005 5 6Google Scholar
Sistonen, JSajantila, ALao O, Corander JBarbujani, GFuselli, S worldwide genetic variation shows high frequency of altered activity variants and no continental structurePharmacogenet Genomics 2007 17 93Google Scholar
Gaedigk, AGotschall, RRForbes, NSSimon, SDKearns, GLLeeder, JSOptimization of cytochrome P450 2D6 (CYP2D6) phenotype assignment using a genotyping algorithm based on allele frequency dataPharmacogenetics 1999 9 669Google Scholar
Kirchheiner, JSchmidt, HTzvetkov, MPharmacokinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to duplicationPharmacogenomics J 2007 7 257Google Scholar
Gasche, YDaali, YFathi, MCodeine intoxication associated with ultrarapid CYP2D6 metabolismN Engl J Med 2004 351 2827Google Scholar
Sallee, FRDeVane, CLFerrell, REFluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiencyJ Child Adol Psychopharmacol 2000 10 27Google Scholar
Voronov, PPrzybylo, HJJagannathan, NApnea in a child after oral codeine: a genetic variant – an ultra-rapid metabolizerPed Anesth 2007 17 684Google Scholar
U.S. Food and Drug Administration 2007 http://www.fda.gov/cder/drug/advisory/codeine.htm
Health Canada. Advisory: Use of Codeine Products by Nursing Mothers 2008 http://www.hc-sc. gc.ca/ahc-asc/media/advisories-avis/_2008/2008_164-eng.php
Gaedigk, ASimon, SDPearce, REBradford, LDKennedy, MJLeeder, JSThe CYP2D6 activity score: translating genotype information into a quantitative measure of phenotypeClin Pharmacol Ther 2007 83 234Google Scholar
Allegaert, KVan, DenAnker, JNVerbesselt, RDemethylation of tramadol in the first months of lifeEur J Clin Pharmacol 2005 61 837Google Scholar
Dematteis, MLallement, GMallaret, MDextromethorphan and dextrorphan in rats: common antitussives – different behavioural profilesFundam Clin Pharmacol 1998 12 526Google Scholar
Nicholson, KLHayes, BABalster, RLEvaluation of the reinforcing properties and phencyclidine-like discriminative stimulus effects of dextromethorphan and dextrorphan in rats and rhesus monkeysPsychopharmacology 1999 146 49Google Scholar
Law, AJWeickert, CSWebster, MJHerman, MMKleinman, JEHarrison, PJExpression of NMDA receptor NR1, NR2A and NR2B subunit mRNAs during development of the human hippocampal formationEur J Neurosci 2003 18 1197Google Scholar
Leeder, JSDevelopmental pharmacogenetics: a general paradigm for application to neonatal pharmacology and toxicologyClin. Pharmacol. Ther 2009 86 687Google Scholar
Leeder, JSUnderstanding the relative roles of pharmacogenetics and ontogeny in pediatric drug development and regulatory scienceJ. Clin. Pharmacol 2010 50 1377Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×