new born pharmacokinetics

Upload: ethan-morgan

Post on 05-Apr-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/2/2019 New Born Pharmacokinetics

    1/20

    Advanced Drug Delivery Reviews 55 (2003) 667686www.elsevier.com/locate/addr

    Pharmacokinetics in the newborna , b*Jane Alcorn , Patrick J. McNamara

    aCollege of Pharmacy and Nutrition, University of Saskatchewan, 110 Science Place, Saskatoon, SK, S7N 5C9, Canada

    bDivision of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA

    Received 11 June 2002; accepted 22 January 2003

    Abstract

    In addition to differences in the pharmacodynamic response in the infant, the dose and the pharmacokinetic processes

    acting upon that dose principally determine the efficacy and/or safety of a therapeutic or inadvertent exposure. At a given

    dose, significant differences in therapeutic efficacy and toxicant susceptibility exist between the newborn and adult.

    Immature pharmacokinetic processes in the newborn predominantly explain such differences. With infant development, the

    physiological and biochemical processes that govern absorption, distribution, metabolism, and excretion undergo significant

    growth and maturational changes. Therefore, any assessment of the safety associated with an exposure must consider the

    impact of these maturational changes on drug pharmacokinetics and response in the developing infant. This paper reviews

    the current data concerning the growth and maturation of the physiological and biochemical factors governing absorption,

    distribution, metabolism, and excretion. The review also provides some insight into how these developmental changes alter

    the efficiency of pharmacokinetics in the infant. Such information may help clarify why dynamic changes in therapeutic

    efficacy and toxicant susceptibility occur through infancy.

    2003 Elsevier Science B.V. All rights reserved.

    Keywords: Development; Exposure; Human; Infant; Newborn; Pharmacokinetics

    Contents

    1. Introduction ............................................................................................................................................................................ 668

    1.1. Pharmacokinetics as a determinant of plasma concentration and response........................ ................... .................... .............. 668

    2. Absorption................... ................... .................... .................... .................... ................... .................... .................... ................. 669

    2.1. Gastrointestinal absorption......................... .................... ................... .................... .................... .................... ................... . 669

    2.1.1. Gastrointestinal secretions................................ .................... .................... ................... .................... .................... .... 6702.1.2. Gastrointestinal motility.......................... ................... .................... .................... ................... .................... .............. 670

    2.1.3. Gastrointestinal metabolism and transport ................... .................... .................... ................... .................... .............. 671

    2.2. Gastrointestinal first-pass effects ................... .................... .................... ................... .................... .................... ................. 671

    3. Distribution........................ .................... .................... .................... ................... .................... .................... ................... ........... 672

    3.1. Body composition and tissue perfusion .................... .................... ................... .................... .................... .................... ....... 672

    3.2. Plasma protein binding .................... .................... .................... ................... .................... .................... ................... ........... 673

    4. Elimination.................. ................... .................... .................... .................... ................... .................... .................... ................. 673

    *Corresponding author. Tel.: 11-306-966-6365; fax: 11-306-966-6377.

    E-mail address: [email protected] (J. Alcorn).

    0169-409X/03/$ see front matter 2003 Elsevier Science B.V. All rights reserved.

    doi:10.1016/S0169-409X(03)00030-9

    mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]
  • 8/2/2019 New Born Pharmacokinetics

    2/20

    668 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    4.1. Hepatic clearance............ ................... .................... .................... ................... .................... .................... .................... ....... 674

    4.1.1. Intrinsic clearance ................. .................... .................... ................... .................... .................... ................... ........... 674

    4.1.1.1. Cytochrome P450 enzyme-mediated metabolism ................... .................... .................... ................... ........... 674

    4.1.1.2. Phase II metabolism............... .................... .................... ................... .................... .................... ................. 676

    4.1.1.3. Consequences of variability in the postnatal maturation of phase I and phase II reactions............... ................. 678

    4.1.2. Hepatic first-pass effects .................. .................... ................... .................... .................... ................... .................... . 6784.2. Renal clearance..... ................... .................... .................... .................... ................... .................... .................... ................. 678

    4.2.1. Glomerular filtration.............. .................... .................... ................... .................... .................... ................... ........... 679

    4.2.2. Renal tubular function .................. ................... .................... .................... ................... .................... .................... .... 679

    5. Conclusions ............................................................................................................................................................................ 680

    References ................... .................... .................... .................... ................... .................... .................... ................... .................... . 680

    1. Introduction affects of postnatal development on drug and toxic-

    ant pharmacokinetics in the newborn and early

    Adult doses, even after adjusted for differences in infancy. The article describes the age-dependent

    body weight, often lead to drastic consequences in changes in the physiological and/or biochemical

    the newborn patient. Functional immaturity of phys- processes governing drug and toxicant phar-

    iological processes and organ function predispose macokinetics. The article then extends these observa-

    newborns to exhibit such disparate responses relative tions to discuss how developmental changes may

    to the adult. With infant maturation, normal develop- lead to significant differences in absorption, dis-

    ment may modify infant response to drug and tribution, metabolism and/or excretion between the

    toxicant exposures. The full impact of developmental newborn, infant and the adult. Such discussion

    immaturity, though, still remains unrealized as ex- should help explain the dynamic changes in thera-

    emplified by the number of well-documented thera- peutic efficacy and toxicant susceptibility that occur

    peutic misjudgments that continue even to this day through infancy.

    [13]. We may mitigate future misjudgments through

    a greater understanding of how development affects 1.1. Pharmacokinetics as a determinant of plasma

    the factors that govern drug and toxicant response in concentration and responsethe newborn and young infant. Such knowledge may

    help ensure safe exposures to therapeutic or inadver- Therapeutic and toxic responses generally corre-

    tent compounds. late well with the plasma concentration of a com-

    Pharmacokinetic and pharmacodynamic processes pound. Because of this correlation, plasma concen-

    contribute to the significant differences in therapeutic trations may best indicate the potential safety and/or

    efficacy and toxicant susceptibility observed between efficacy of the compound in the newborn and young

    newborns, young infants and adults. With postnatal infant. This presumes that the pharmacological or

    development, growth and functional maturation of toxic response is direct (i.e., related to corresponding

    the biochemical and physiological factors governing serum concentrations), expected (i.e., similar to adult

    pharmacokinetics may alter the processes of absorp- response) and quantitatively similar to adults (i.e.,

    tion, distribution, metabolism and excretion. As well, similar pharmacodynamic parameters). A more dif-these developmental changes proceed along a con- ficult situation arises if the pharmacological or toxic

    tinuum, at different rates and patterns, resulting in response is indirect (i.e., unrelated to serum con-

    tremendous interindividual variability in infant phar- centrations), novel (a function of the developing

    macokinetics. The dynamic and highly variable neonatal physiology/biochemistry) and quantitative-

    character of postnatal maturation of infant phar- ly dissimilar to adults. Whether pharmacodynamic

    macokinetic and pharmacodynamic processes may differences exist between pediatric population groups

    have significant consequences on the way newborns and adults is largely unknown. Given this presump-

    and infants respond to and deal with drugs. tion, two factors govern plasma concentrations, the

    As its principal goal, this review discusses the size of the dose and the pharmacokinetic processes

  • 8/2/2019 New Born Pharmacokinetics

    3/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 669

    of absorption, distribution, metabolism and excretion exhibit differences in toxicant susceptibility or thera-

    acting upon that dose. The following equations peutic efficacy.

    illustrate the influence of dose and pharmacokinetic In general, the combined effects of maturation of

    processes on plasma concentrations of a compound, each pharmacokinetic process on the plasma levels

    administered as either a multiple oral dose (Eq. (1)) of a given compound are not well understood.or as a single oral dose (Eq. (2)). Premature birth (gestational age ,36 weeks) and

    underlying pathophysiology further complicate theFD relationship between plasma concentrations and re-]

    t sponse and the age-related changes in phar-]]C5 (multiple dose) (1)Cl

    S macokinetics. Premature infants exhibit more pro-

    nounced anatomical and functional immaturity of theor organs involved in pharmacokinetic processes. The

    extent to which premature infants differ from theCl SFk D

    a ]2 t 2 k tS D s da full-term infant correlates directly with the degree ofV]]]S DC5 e 2 e (single dose)dt V k 2 ks dd a e prematurity [4]. This enhanced immaturity, as wellas any underlying disease state(s), may impede

    (2) normal postnatal development of the processes of absorption, distribution and elimination. How thesewhere C is the average steady state plasma con-

    and other factors may contribute to age-dependentcentration; F is the bioavailability; D is the dose; tpharmacokinetics in newborns and infants requiresis the dosing interval; Cl is the systemic clearance;

    S

    consideration. The proceeding discussion will sum-C is the plasma concentration at any time, t; k ist a

    marize the available literature on the influence ofthe absorption rate constant; k is the elimination ratee

    postnatal maturation on drug absorption, distribution,constant and is equal to Cl /V ; V is the volume ofS d dmetabolism and excretion principally in the full-termdistribution; and t is time.infant.According to Eqs. (1) and (2), larger doses (D)

    result in higher plasma concentrations of an adminis-

    tered compound. As well, these equations clearly

    2. Absorptionillustrate the importance of absorption (indicated inthe F and k terms), distribution (indicated in the V

    a d

    Age-related differences in absorption relate toterm), and metabolism and excretion (indicated indevelopmental changes in those factors governingthe Cl and k terms) as determinants of plasmaS epassive or carrier-mediated transport across theconcentrations in the newborn and young infant.absorptive barrier. Systemic bioavailability becomesRapid changes in body size and composition, organan important consideration when compounds aresize and function, and maturation of the underlyingabsorbed from extravascular administration sites.physiological and biochemical processes that governThe absorptive characteristics of the compound andabsorption, distribution, metabolism, and excretionthe possible influence of first-pass effects may limitcharacterize the immediate postnatal period. Thesethe systemic bioavailability of the compound. Thisdevelopmental changes cause major age-related

    may lead to lower plasma concentrations of thechanges in drug absorption, distribution and elimina-compound and reduced exposures.tion (metabolism and excretion), which may have a

    significant impact on plasma concentrations and the2.1. Gastrointestinal absorptionresultant exposure outcomes. Additionally, matura-

    tion is a dynamic process influenced by a plethora ofTable 1 summarizes the age-dependent anatomicalgenetic and environmental factors. The rate and

    and physiological factors that may influence the ratepattern of maturation of each pharmacokinetic pro-and/ or extent of gastrointestinal absorption. De-cess may vary greatly among infants. This may resultvelopmental changes in one or any combination ofin marked interindividual variability in phar-these factors may explain the differences in absorp-macokinetics such that infants of similar age may

  • 8/2/2019 New Born Pharmacokinetics

    4/20

    670 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    Table 1a

    Age-dependent factors affecting gastrointestinal absorption and the resultant pharmacokinetic outcome relative to adult levels

    Newborn Neonate Infant

    (full-term) (1 day1 month) (1 month2 years)

    Physiological factorGastric pH 13 .5 |Adult

    Gastric emptying time Reduced (variable) Reduced (variable) Increasedb b

    Intestinal surface area Reduced Reduced |Adult

    Intestinal transit time Reduced Reduced Increased

    Pancreatic and biliary function Very immature Immature |Adult

    Bacterial flora Very immature Immature Immature

    Enzyme/transporter activity Very immature Immature Approaching adult

    Pharmacokinetic outcome

    Rate and extent of absorption Variable Variable $Adult

    Gastrointestinal first-pass effects Very reduced Reduced Approaching adult

    aAdapted from Besunder et al. [7].

    b

    From Ref. [8].

    tive characteristics between newborns and young pH in the newborn and young infant may involve

    infants relative to the adult. The developmental enhanced bioavailability of weakly basic compounds,

    pattern of these processes may be highly variable and but reduced bioavailabilities of weakly acidic com-

    environmental factors (i.e., diet, concomitant drugs) pounds. This may explain the increased bioavail-

    [5], genetic factors and underlying pathophysiology ability of ampicillin and penicillin G (basic drugs)

    (electrolyte abnormalities, endocrinopathies, CNS [1719] and decreased bioavailability of phenobarbi-

    disorders, gastrointestinal disease) [6] may poten- tal (acidic drug) [19,20] observed in young infants.

    tially influence their postnatal developmental pattern. Certain compounds require pancreatic exocrine

    In general, newborns exhibit a slower rate of and biliary function for adequate absorption. New-

    absorption [6]. Age-dependent differences in the borns have immature pancreatic and biliary functionextent of absorption (bioavailability) remain largely at birth [21]. The levels of most pancreatic enzymes

    unknown [8]. However, bioavailability (the fraction are significantly reduced [22], and bile formation,

    of parent compound reaching the systemic circula- bile acid pool size (50% adult values), bile acid

    tion) will likely change with postnatal age due to synthesis and metabolism, and bile acid intestinal

    maturation of the processes governing absorption. absorption are all reduced in the newborn [2325].

    Pancreatic and biliary function rapidly develop in the

    2.1.1. Gastrointestinal secretions postnatal period [22,26]. A deficiency of bile salts

    Gastrointestinal pH affects the absorption of weak- and pancreatic enzymes may result in a reduction in

    ly acidic and basic organic compounds. At birth, the bioavailability of those drugs that require

    newborns have an alkaline gastric pH (pH 68) solubilization or intraluminal hydrolysis (i.e., pro-

    [9,10]. Gastric acid production increases over the drug esters) for adequate absorption.next 2448 h to achieve adult pH levels (pH 13)

    [1113]. Following this initial burst of hydrochloric 2.1.2. Gastrointestinal motility

    acid secretion, gastric acid production declines and Gastrointestinal motility may affect the rate and/

    gastric acidity remains relatively low in the first or extent of drug absorption. In general, newborns

    months of life [11,14]. Postnatal increases in gastric exhibit delayed gastric emptying rates and prolonged

    acid production generally correlate with postnatal intestinal transit times relative to the adult [27,28].

    age [15] and, on a per kg basis, adult levels are The full-term newborn infant demonstrates quali-

    approached by 2 years of age [16]. tatively similar gastrointestinal motility patterns with

    The pharmacokinetic consequences of high gastric the adult, but premature infants exhibit disorganized

  • 8/2/2019 New Born Pharmacokinetics

    5/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 671

    and inefficient motility patterns [29,30]. In general, The developmental maturation of gastrointestinal

    feeding triggers the postnatal development of gas- Phase II conjugation enzymes [4247] remains

    trointestinal motility [5]. Reduced gastrointestinal unknown. However, b-glucuronidase activity of the

    motility may have variable and unpredictable effects infant small intestine has been reported to exceed

    on drug bioavailability in newborns and young adult activities by as much as 7-fold [48]. Thisinfants. In general terms, delayed gastric emptying enhanced b-glucuronidase activity may enable re-

    may reduce the rate of drug absorption since the absorption of glucuronide drug conjugates. For drugs

    small intestinal mucosa acts as the principal absorp- that undergo enterohepatic recirculation (i.e.,

    tive site for most drugs. Alternatively, slower intesti- chloramphenicol, indomethacin) b-glucuronidase ac-

    nal transit times may improve drug bioavailability tivity enhances their bioavailability.

    due to longer retention times in the small intestine. The adult intestinal tract functionally expresses

    The exact effect of developmental maturation of various members of the ATP-binding cassette and

    gastrointestinal motility on drug bioavailability de- solute carrier transporter families [49,50]. These

    pends upon the physico-chemical properties of the transporters may have an important impact on drug

    drug and its interaction with the anatomical and absorption and bioavailability in the small intestine

    physiological factors of the gastrointestinal tract. [51], but their exact role remains largely unknown.

    The literature provides evidence of postnatal matura-

    2.1.3. Gastrointestinal metabolism and transport tion of transport protein activities in other organ

    Bacterial flora, principally concentrated in the systems [5258] suggesting gastrointestinal transpor-

    ileum and colon [31], may influence the extent of ter function may also undergo a postnatal maturation.

    drug absorption due to its influence on gastrointesti-

    nal motility and ability to metabolize compounds 2.2. Gastrointestinal first-pass effects

    [32]. At birth, infant gastrointestinal flora is very

    immature and little information is available on the The gastrointestinal tract may play an important

    effect of postnatal maturation of bacterial flora on role in the first pass metabolism of an orally adminis-

    bioavailability [32,33]. In general, the bacterial flora tered compound [51]. Consequently, developmental

    of the infant gastrointestinal tract approaches adult maturation of gastrointestinal metabolic and transport

    populations by 4 years of age [34]. function may have significant consequences in gas-The proximal small intestine acts as the principal trointestinal first-pass effects and oral bioavailability.

    absorptive site and site for significant first-pass In adults, some drugs undergo extensive metabolism

    effects for many orally administered compounds. In in the gastrointestinal tract with a concomitant

    the adult, the small intestinal mucosa functionally marked reduction in their oral bioavailability [59

    expresses a limited number of phase I and phase II 61]. Immature gastrointestinal metabolic reactions

    enzymes and their expression has led to significant may result in improved oral bioavailability as gas-

    inter- and intra-individual variability in oral bioavail- trointestinal first-pass metabolism has less influence

    ability [35]. Cytochrome P450 (CYP) 3A4 is the on the extent of absorption of such drugs. Converse-

    predominant CYP enzyme expressed in enterocytes ly, some drugs are dependent upon carrier-mediated

    [36,37], and CYP2C has the second highest expres- uptake systems in the intestinal mucosa for their

    sion levels [37].Very low activity levels for CYP1A1 efficient absorption [62]. Immature development of[38] and CYP2D6 [35,39] were detected in intestinal transport function may lead to significantly reduced

    microsomes. The maturation of CYP enzymes in the oral bioavailability.

    intestinal mucosa remains largely uninvestigated. Some transporter proteins expressed in the intesti-

    One study reported significantly lower CYP3A4 nal mucosa promote the active extrusion of drug

    activity levels in intestinal microsomes from infants from the enterocyte back into the lumen of the

    aged 03 months and a developmental increase in gastrointestinal tract after its absorption (i.e., MDR1)

    activity with age [40]. As well, CYP3A7, the fetal [63,64]. These transporter proteins compete with

    hepatic form of CYP3A, appears to lack expression absorption processes and may decrease the rate of

    in extrahepatic tissues [41]. absorption and, potentially, the oral bioavailability.

  • 8/2/2019 New Born Pharmacokinetics

    6/20

    672 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    The interplay between CYP3A4 and the active efflux postnatal period [65], while total body fat increases

    transporter, MDR1, illustrates this concept [63,64]. progressively in the first months of life [66].

    Again, immature development of these active efflux Age-related changes in total body water are pri-

    processes may result in enhanced oral bioavailability. marily attributed to decreases in the relative per-

    The exact effect of postnatal maturation on oral centage of extracellular water [65,67]. Extensivebioavailability will depend upon the interaction of tissue binding or partitioning into fat contribute to

    the principal factors influencing bioavailability large V values. Polar compounds generally exhibit Vd d

    (physico-chemical properties of the compound, equivalent to total body water or blood volumes.

    physiology and anatomy of the gastrointestinal tract, Hence, age-related changes in fat, muscle and total

    metabolic enzymes, transport processes) [51] and the body water composition may produce significant

    degree of their postnatal maturation. quantitative changes in V and plasma concentrations.d

    In newborns, the high relative proportion of total

    body water and low proportion of fat results in a

    general increase in V for water-soluble compoundsd3. Distribution

    and a lower V for fat-soluble drugs relative to adults.d

    Key pharmacokinetic parameters (i.e., clearancePostnatal changes in body composition, extent of and volume of distribution) are often normalized

    binding to plasma proteins and tissue components,according to total body weight or body surface area.

    and hemodynamic factors (cardiac output, tissueTherefore, it is critical to understand the develop-

    perfusion and membrane permeability) may altermental changes in these body indices with postnatal

    distribution characteristics in the developing infant.age, which is depicted in Fig. 1. While both total

    The apparent volume of distribution (V ) provides ad body weight and surface area rise steadily during the

    useful marker to assess age-related changes in drugfirst year of life, a considerable change in their ratio

    distribution.occurs during the initial 3 months of life.

    Developmental changes in V may also relate tod

    3.1. Body composition and tissue perfusion postnatal enhancements in cardiac output, organ

    blood flows and tissue perfusion, changes in mem-

    Body composition may significantly affect drug brane permeabilities [7072] and maturation ofV . Changes in body composition correlate with both carrier-mediated transport systems [5558,7375],

    d

    gestational and postnatal age. Table 2 illustrates total and changes in tissue binding affinities or capacities

    body water, total protein and total fat content in the since newborns and young infants have significantly

    newborn, during infancy and in the adult stages.

    Total body water decreases significantly in the early

    Table 2

    Developmental changes in body composition (reported as aa

    percentage of total body mass)

    Age Body Water Protein Fat

    mass

    (kg)

    Newborn: full-term 3.5 74 11 14

    4 months 7.0 61.5 11.5 27

    12 months 10.5 60.5 15 24.5Fig. 1. Age-related changes in body weight, body surface areab

    Adult 70 5560 (BSA) and the ratio of body weight to BSA. Data from the Center

    aAdapted from Geigy Scientific Tables [66]. for National Health Statistics at the CDC [68] and Taketomo et al.

    bObesity decreases the percentage of total body water. [69].

  • 8/2/2019 New Born Pharmacokinetics

    7/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 673

    greater liver, kidney and brain masses relative to

    total body mass [66,67,76].

    3.2. Plasma protein binding

    Postnatal development of plasma protein binding

    may affect both the distribution and elimination of

    compounds in the newborn and young infant. In

    general, newborns and young infants exhibit larger

    unbound fractions of a compound relative to the

    adult. This may result in enhanced distribution intoFig. 2. General pattern of ontogeny for albumin (ALB) and alpha

    tissues and a larger V . Age-related differences ind 1-acid glycoprotein (AAG) in newborns and infants. Data adapted

    plasma protein binding affinity, plasma protein con- from [86] and are expressed as fraction of adult serum con-centration (mg/dl).centrations and availability of competing endogenous

    compounds largely explain the differences in the

    extent of binding.

    Compounds bind principally to albumin and a -acid glycoprotein plasma concentrations and the1

    alpha -acid glycoprotein. Albumin is the major fraction bound in the infant relative to the adult [86].1

    plasma protein [77], and concentrations of alpha - Several endogenous substances (i.e., bilirubin,1acid glycoprotein, an acute phase reactant protein fatty acids) may compete for plasma protein binding

    [78], fluctuate significantly in response to various sites [82,8789]. This competition may further re-

    diseases, trauma or chemical insult [79,80]. In gener- duce the bound fraction of a compound in the

    al, the newborn may exhibit lower binding affinities newborn, or cause displacement of the endogenous

    of compounds (i.e., penicillin, phenobarbital, pheny- molecule from its protein binding sites. For instance

    toin and theophylline) [81] to albumin and a -acid hyperbilirubinemia reduces the binding of the acidic1

    glycoprotein [8284]. High unbound fractions may drugs ampicillin, penicillin, phenobarbital and

    lead to significantly larger V values, and enhanced phenytoin [81,90]. Conversely, displacement of bil-d

    renal clearance by glomerular filtration and hepatic irubin by drugs (i.e., sulfonamides) may enhance theclearance of low extraction ratio compounds in the risk for bilirubin encephalopathy [7]. Hepatic and

    newborn. renal disease, hypoproteinemia due to malnutrition,

    Developmental changes in plasma protein con- cystic fibrosis, burns, malignant neoplasms, surgery,

    centrations have the most significant affect on the trauma and acidosis may further decrease plasma

    extent of plasma protein binding in the young infant. protein drug binding due to decreased protein syn-

    Total plasma protein levels are lower in the newborn thesis or competition for binding.

    relative to the adult [85]. Lower plasma protein

    levels reduce the plasma protein binding capacity in

    the newborn. Fig. 2 illustrates the postnatal increase

    in albumin and alpha -acid glycoprotein concen- 4. Elimination1

    trations relative to adult levels. A strong correlationexists between the postnatal increase in plasma Systemic clearance provides a measure of the

    albumin concentrations and the fraction bound [86]. efficiency of elimination and is often the most

    This suggests adult binding characteristics (i.e., important pharmacokinetic determinant of plasma

    unbound fraction) for a given compound and the concentrations and resultant response (see Eq. (1)).

    ratio of infant and adult albumin concentrations may In general, hepatic and/or renal elimination path-

    provide an estimate of infant unbound fractions for ways effect the removal of most compounds from the

    that compound [86]. On the other hand, a weak body. These pathways are generally underdeveloped

    correlation exists between the postnatal increase in and inefficient in the newborn. The various pathways

  • 8/2/2019 New Born Pharmacokinetics

    8/20

    674 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    of elimination mature at different rates and patterns thione conjugation, and acetylation. Since most

    of development and maturation to adult levels (ad- compounds are eliminated by more than one meta-

    justed for body weight differences) is generally bolic pathway, postnatal changes in the efficiency of

    achieved after the first year of postnatal life. Recent Phase I and Phase II reactions, differences in their

    in vitro and in vivo probe substrate data have rate and pattern of development, and changes in theprovided important information on the maturation hepatocellular distribution and expression of Phase I

    characteristics of hepatic and renal elimination mech- and Phase II enzymes [104] may have a significant

    anisms (reviewed in Alcorn and McNamara, 2002) impact on the qualitative and quantitative charac-

    [91]. This data provides the basis for the proceeding teristics of hepatic elimination in the newborn and

    discussion. Additionally, for more in depth discus- developing infant. To predict the exact nature of

    sion of the maturation of systemic clearance mecha- these consequences requires an understanding of the

    nisms the reader is referred to excellent reviews by postnatal maturation of the individual hepatic meta-

    Hakkola et al. [92], Ring et al. [93], Gow et al. [94], bolic pathways that mediate drug and toxicant re-

    McCarver and Hines [95], Hines and McCarver [96], moval from the body.

    de Wildt et al. [97], and Hayton [98].4.1.1.1. Cytochrome P450 enzyme-mediated metabo-

    4.1. Hepatic clearance lism

    The CYP enzymes represent a superfamily of

    Hepatic blood flow, plasma protein binding and heme-containing enzymes [105]. CYP1A2, CYP2A6,

    intrinsic clearance (defined as the maximal en- CYP2B6, CYP2Cs, CYP2D6, CYP2E1, and

    zymatic or transport capacity of the liver) constitute CYP3A4/7 comprise the principal CYP enzymes

    the physiological determinants of hepatic clearance important in drug and toxicant metabolism [106

    [99,100]. Each of these determinants undergoes 108]. The rate and pattern of postnatal CYP enzyme

    significant postnatal changes, and their maturation development may have a significant impact on

    results in an enhanced capacity for hepatic elimina- therapeutic efficacy and toxicant susceptibility in the

    tion of compounds with advancing postnatal age. newborn and developing infant. Interindividual dif-

    ferences in their developmental patterns, genetic

    4.1.1. Intrinsic clearance polymorphisms, and their induction/ inhibition po-Intrinsic clearance processes principally govern tential further complicate the role of CYP enzyme

    the capacity of newborns to eliminate drug by the maturation on pharmacokinetics in the newborn and

    liver. Although hepatocellular transport and biliary young infant.

    excretion processes contribute to intrinsic clearance The postnatal maturation of CYP enzymes is

    and are deficient at birth [74,101,102], hepatic evidenced in numerous literature reports of short-

    biotransformation processes have the greatest impact ening half-lives and enhanced hepatic elimination of

    on hepatic drug elimination in the developing infant. drugs in developing infants [109,110]. These studies

    Phase I and Phase II reactions principally mediate suggest hepatic metabolic pathways undergo rapid

    the metabolism of compounds in the developing postnatal development. Recently, in vitro studies

    infant. Often a compound undergoes sequential have examined the maturation of individual CYP

    metabolism with Phase I metabolic reactions preced- enzymes in age-dependent fetal and infant hepaticing Phase II metabolism [103]. microsomes [111115]. These studies corroborate

    Of the phase I reactions, cytochrome P450 (CYP) the findings of the in vivo studies and have further

    enzymes have the most important role in the elimina- elucidated the maturation characteristics of individ-

    tion of most compounds. The postnatal maturation of ual CYP enzymes. As well, these studies have shown

    other Phase I enzymes, such as the alcohol and the CYP enzymes mature at characteristic rates and

    aldehyde dehydrogenases, esterases and the flavin- patterns of development and may be grouped accord-

    containing monooxygenases, are reviewed in Hines ing to their general developmental pattern of activity

    and McCarver, 2002 [96]. Important phase II re- [116]. Fig. 3 highlights the general pattern of CYP

    actions include glucuronidation, sulfation, gluta- enzyme development as a fraction of adult levels

  • 8/2/2019 New Born Pharmacokinetics

    9/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 675

    biotransformation and, in general, CYP enzyme-me-

    diated metabolism improves with postnatal age and

    generally approaches adult levels only after the first

    year of life [117,120]. Table 3 summarizes the

    maturation of individual CYP enzyme activity levelsbased upon in vitro determinations of CYP enzyme

    activity in fetal, infant and adult age group hepatic

    microsomes [111115].

    4.1.1.1.1. Maturation of individual CYP

    enzymes Fetal hepatic microsomes exhibit negligible

    CYP1A2 enzyme activity [107,121,122]. CYP1A2

    enzyme activity remains very low after birth and

    significant in vitro activity is detected only by 13

    months of age [112]. By the first year of lifeFig. 3. General pattern of postnatal development of the hepatic

    CYP1A2 enzyme activity levels are only 50% adultclearance pathways in newborns and infants. Data adapted from

    values and mature to adult activity levels sometime[91] and are expressed as fraction of adult clearance values after a year of age [112,120]. This pattern of(ml/min/kg). The general classification was adapted from Cres-teil, 1998 [116]. (CYP, cytochrome P450 enzyme; UGT, Uridine development explains the long half-life and low59-diphosphate-glucronosyltransferase; ST, Sulfotransferase; NAT, systemic clearance values of theophylline in the

    N-acetyltransferase; GST, Glutathione-S-transferase).newborn [123]. Immature CYP1A2 enzyme develop-

    ment prevents the biotransformation of theophylline

    based upon enzyme activity values (normalized to resulting in prolonged half-lives in the newborn and

    mg microsomal protein) from age-specific fetal, infant. Significant increases in theophylline systemic

    neonatal and infant hepatic microsomes [116]. clearance values are observed only after 13 months

    In vitro assessments have revealed significantly of age [123,124]. This pattern of theophylline clear-

    lower levels of CYP enzyme protein and activity in ance with advancing postnatal age reflects the post-

    the fetus (total CYP enzyme protein levels are one- natal development of in vitro CYP1A2 enzyme

    third adult levels) [107,117,118]. These data are activity in the infant.consistent with literature reports demonstrating the Fetal livers fail to express CYP2A6 and CYP2B6

    ability of the fetal liver to metabolize a variety of enzyme activity [107,122]. Otherwise, the postnatal

    drug substrates [116,119]. For most CYP enzymes, development of CYP2A6 and CYP2B6 remains

    though, fetal activity levels are only a small fraction largely unknown. Both CYP enzymes likely achieve

    of adult activity levels, and parturition triggers their adult capacities only after the first year of life [120].

    postnatal development [9294,116]. Consequently, Fetal and newborn (,1 week of age) livers

    the newborn has a limited capacity for hepatic demonstrate very limited CYP2C enzyme activity

    Table 3

    In vitro cytochrome P450 (CYP) enzyme activity in age-specific fetal and infant hepatic microsomes as a fraction of adult activity (nmol21 21

    min mg microsomal protein )CYP Fraction of adult activity

    enzymeFetus ,24 h 17 d 828 d 13 m 312 m 115 y

    1A2 0.05 0.12 0.10 0.20 0.39 0.46 1.10

    2C 0.02 0.03 0.42 0.29

    2D6 0.04 0.04 0.09 0.24

    2E1 0.21 0.31 0.36 0.46 0.39 0.80

    3A4 0.03 0.08 0.13 0.29 0.34 0.43 1.08

    3A7 5 9.5 13 6 3 2

    Adapted from Refs. [111115].

  • 8/2/2019 New Born Pharmacokinetics

    10/20

    676 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    [118,125]. Within the first month of postnatal life, activity and express limited CYP3A4 enzyme activi-

    CYP2C enzyme activity surges to 50% adult levels ty (|10% adult levels) [111,122]. CYP3A7 enzyme

    [114,117]. After this surge of activity, CYP2C activity levels peak 1 week after parturition, then

    enzyme activity levels decline slightly for the first declines significantly during the first year of life

    year of life and adult levels are reached sometime [111,120]. Adult livers may express only 10% fetalafter 1 year of age [114,117,120]. As a substrate of levels [111,132]. During the postnatal period, activi-

    the CYP2C enzyme subfamily, diazepam metabolite ty levels of CYP3A4 enzyme increase concomitantly

    urinary levels are consistent with the in vitro pattern with the decreases in CYP3A7 enzyme activity

    of CYP2C enzyme development. Newborns exhibit [111]. CYP3A4 enzyme activity reaches 3040%

    very low urinary diazepam metabolite levels [114]. adult levels by 1 month of age and adult levels by 1

    However, diazepam urinary metabolite levels in- year [111,120]. The pharmacokinetic consequences

    crease significantly in infants greater than 1-week- of this postnatal developmental switch from

    old [114]. Thereafter, metabolite levels remain rela- CYP3A7 to CYP3A4 remain largely unknown since

    tively stable in children up to 5 years of age [114]. the substrate profile of CYP3A7 enzyme has re-

    Fetal livers may express very low levels of ceived limited investigation. However, some studies

    CYP2D6 enzyme activity [107,113]. A dramatic suggest newborns and adults will exhibit significant

    increase in CYP2D6 activity occurs in the immediate differences in their capacity to eliminate known

    postpartum period. By the first month of life, CYP3A4 enzyme substrates. For example, premature

    CYP2D6 enzyme activity levels reach |30% adult and full-term newborns eliminate the CYP3A4 en-

    levels [113], and CYP2D6 maturation may be com- zyme substrate, midazolam, with poor efficiency

    pleted by 1 year of age [120]. In vivo assessment of [133]. A 5-fold increase in the elimination efficiency

    the hepatic clearance of CYP2D6 substrates is of midazolam occurs by 3 months of age [134].

    lacking in the newborn and young infant. These data suggest midazolam is not an efficient

    Fetal hepatic microsomes may express low levels CYP3A7 enzyme substrate.

    of CYP2E1 enzyme activity [126]. Parturition trig-

    gers a dramatic increase in CYP2E1 enzyme activity 4.1.1.2. Phase II metabolism

    within the first 24 h of life [115]. CYP2E1 enzyme Phase II or conjugation reactions contribute sig-

    activity levels achieve 50% adult levels by 13 nificantly to the elimination of a wide variety ofmonths of age and development is essentially com- exogenous and endogenous compounds. Glucu-

    plete after 1 year of age [115]. ronidation, sulfation, acetylation, glutathione conju-

    CYP3A subfamily is the most abundantly ex- gation, comprise the most important Phase II path-

    pressed CYP enzyme in the liver [106,127]. ways in drug and toxicant metabolism. In general,

    CYP3A4 enzyme is the principal enzyme of the adult changes in the expression patterns of the different

    liver [106], while fetal livers predominantly express Phase II enzymes or changes in their catalytic

    CYP3A7 enzyme [111,128,129]. Although CYP3A4 efficiency may occur with development. Such

    and CYP3A7 enzymes exhibit 95% similarity in their changes may have important consequences on the

    nucleotide sequences [130], important differences in elimination of compounds in the newborn and young

    substrate specificities exist between these two infant. In general, inefficient conjugation capacity of

    CYP3A enzymes [107,111,131]. Few studies have the newborn will result in a significant reduction inexamined the substrate profile of CYP3A7 enzyme. the ability of the newborn to eliminate both exogen-

    The ability of fetal livers to metabolize a wide ous and endogenous compounds. Table 4 and Fig. 3

    variety of substrates [119] suggests CYP3A7 enzyme summarize the known maturation patterns of im-

    also may metabolize a wide range of substrates and portant Phase II enzymes. For a more in depth

    demonstrate some substrate overlap with CYP3A4 discussion of the development of Phase II metabolic

    enzyme. pathways, the reader is referred to the review by

    Total CYP3A enzyme protein levels remain rela- McCarver and Hines [95].

    tively constant throughout development [111]. Fetal 4.1.1.2.1. Maturation of individual conjugation

    livers demonstrate high levels of CYP3A7 enzyme reactions The uridine 59-diphosphate-glucrono-

  • 8/2/2019 New Born Pharmacokinetics

    11/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 677

    Table 4 capacity to eliminate morphine, and adult levels wereMaturation patterns of phase II enzymes achieved by 630 months of age [146,147].Phase II enzyme Maturation pattern Sulfotransferases (ST) consist of a number of

    individual enzymes and have substrate specificitiesUGT Fetal livers exhibit limited enzyme activity;

    activity |25% adult levels at 3 months; that demonstrate significant overlap with the UGTmaturation is isoforms specific; adult enzymes [148]. Although changes in activity of theactivity levels achieved by 630 months. individual ST enzymes with development occur, the

    data on their development is limited and confusing.ST Fetal livers exhibit significant activity;

    In general, fetal, newborn and infant livers expressmaturation is isoform specific.significant ST activity, and sulfate conjugation is a

    GST Fetal livers exhibit significant activity; relatively efficient pathway at birth [149151]. Con-maturation is isoform specific; total sequently, the newborn and young infant may elimi-activity remains stable throughout infancy.

    nate ST enzyme substrates very efficiently. For

    instance, the ST and UGT enzyme substrate ritod-NAT Fetal livers exhibit low activity; lowactivity at birth through the first months of rine, a b -adrenoceptor agonist, underwent extensive

    2

    life; adult levels achieved after 1 year of age. sulfate conjugation in infants [149]. The study

    demonstrated no age-related differences in the over-UGT, Uridine 59-diphosphate-glucronosyltransferase; ST,Sulfotransferase; NAT, N-acetyltransferase; GST, Glutathione-S- all systemic clearance of ritodrine, only quantitativetransferase. differences in metabolite levels (glucuronide conju-

    gates and sulphate conjugates) between infants and

    adults [149].

    syltransferases (UGT) enzymes consist of two Glutathione-S-transferases (GST) represent a

    families, UGT1 and UGT2, containing more than 18 superfamily of dimeric enzymes responsible for the

    different enzymes [97,135]. More than one UGT detoxification of a number of potentially toxic drug

    enzyme may participate in the metabolism of a single or drug metabolites [152]. Five different subunit

    substrate [136,137], and the maturation of UGT classes (m, a, u, p and z) of GST enzymes have

    enzyme capacity is isoform specific [97,138]. Conse- been classified [152,153]. As with the ST enzymes,

    quently, the developmental rate and pattern of in- the GST enzymes demonstrate age-related expressiondividual UGT enzymes may explain the tremendous of individual enzymes in the liver [154156]. For

    variability reported in the glucuronidation capacity of example, preterm newborn livers exhibited 60%

    newborns and infants. In their review on the de- greater activity towards chloramphenicol than fetal

    velopmental maturation of glucuronidation capacity livers, but showed similar activity levels towards

    in the infant, de Wildt et al. suggest the clinical chlorodinitrobenzene [157]. However, the literature

    relevance of the development of individual UGT remains sparse and presents conflicting information

    enzyme capacity in infants remains unclear [97]. with respect to individual enzyme development

    As a whole, newborns and young infants demon- [156,157]. In general, though, GST activity is rela-

    strate inefficient glucuronidation capacity relative to tively well-developed in the newborn and infant, and

    the adult [139141]. Fetal livers exhibit limited UGT total GST activity may remain relatively stable

    enzyme activity [142,143]. Fetal hepatic microsomes throughout development [158]. The clinical signifi-(1527 weeks gestation) catalyzed morphine glucu- cance of the quantitative and qualitative differences

    ronidation at only 1020% the efficiency of adult in the developmental expression of individual GST

    hepatic microsomes [144,145]. Parturition triggers an enzymes remains unknown, but may have important

    increase in UGT enzyme activity and UGT enzyme implications in toxicant susceptibility.

    activity achieves |25% adult levels by 3 months of N-acetyltransferases (NAT) consist of two en-

    age [143]. The in vivo postnatal elimination pattern zymes, NAT1 and NAT2, and NAT2 demonstrates

    of the UGT enzyme substrate, morphine, is con- polymorphic activity [159161]. Very limited data

    sistent with the in vitro studies. Premature and full- exists on the developmental expression of NAT1 and

    term infants have a markedly reduced and variable NAT2 enzyme activity. Fetal livers express activity

  • 8/2/2019 New Born Pharmacokinetics

    12/20

    678 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    towards several NAT enzyme substrates, but at much assessment of elimination capacity in the newborn

    lower levels than the adult [47,162]. Consequently, and young infant.

    the newborn exhibits a limited capacity to acetylate

    substrates. The acetylation status of the infant may 4.1.2. Hepatic first-pass effects

    reflect adult levels only well past the first year of life For many drugs subject to first-pass effects,[163165]. hepatic metabolism results in a significant reduction

    in the oral bioavailability of a compound. Postnatal4.1.1.3. Consequences of variability in the postnatal maturation of metabolic enzyme pathways and hepa-

    maturation of phase I and phase II reactions tocellular transport systems may result in significant

    Many compounds undergo multiple routes of differences in the oral bioavailability of compounds

    metabolism. Quantitative and qualitative differences in the newborn and young infant relative to the adult.

    in the developmental expression profiles of metabolic For all orally administered compounds, plasma pro-

    pathways of the liver may modify the rate and tein binding and intrinsic clearance determine the

    pattern with which newborns and infants eliminate extent of parent drug bioavailability [99]. Newborns

    compounds throughout development. These inter- generally exhibit lower plasma protein binding ca-

    individual differences may lead to altered metabolite pacities. Higher unbound fractions of an absorbed

    profiles as the relative contribution of the different compound may theoretically enhance oral clearance

    routes of metabolism may vary with infant age. and result in lower oral bioavailability of the com-

    Differences in metabolite profiles become significant pound. For most drugs, intrinsic clearance has the

    when a particular metabolite has pharmacological or most important effect on bioavailability. At birth,

    toxicological activity. Consequently, the rate, pattern immature Phase I and Phase II metabolic enzyme

    and extent of metabolic pathway maturation are pathways and hepatocellular transport processes may

    important considerations in the pharmacokinetic significantly reduce the extent of first-pass hepatic

    consequences of postnatal maturation of hepatic metabolism of an absorbed compound. Inefficient

    clearance pathways. Interindividual differences in the hepatic metabolism in the newborn may cause

    rates and patterns of individual elimination pathway enhanced oral bioavailabilities relative to the adult.

    maturation may largely explain the tremendous Maturation of the hepatic metabolic pathways will

    interindividual variation observed in the capacity of result in age-related reductions in oral bioavail-newborns and infants to eliminate drugs. Superim- ability. As with hepatic clearance, interindividual

    posed upon the interindividual variability in metabol- differences in the rate and pattern of metabolic

    ic enzyme maturation is the influence of metabolic enzyme pathway maturation may cause significant

    enzyme induction and/ or inhibition. In utero and/ or interindividual variation in oral bioavailability during

    postnatal exposure to certain exogenous or endogen- postnatal development. Hence, postnatal maturation

    ous compounds may cause rapid enzyme induction of hepatic metabolism may greatly influence thera-

    or inhibition in the fetus, newborn and infant [166]. peutic efficacy and toxicant susceptibility because

    This will further exacerbate the variable rate and hepatic metabolism may determine the both the oral

    pattern of enzyme maturation. For example, new- bioavailability of a compound and the efficiency with

    borns treated concomitantly with barbiturates, a which the newborn or young infant may remove that

    CYP2C inducer [114], exhibited a marked reduction compound from the body.in diazepam (a CYP2C substrate) half-lives (t 5

    1 / 2

    1861 h) as compared with newborns treated with 4.2. Renal clearance

    diazepam alone (t 53162 h) [114,167]. Further-1 / 2

    more genetic polymorphisms in CYP2D6, CYP2C9, Renal clearance mechanisms include glomerular

    CYP2C19, CYP2E1, UGT and NAT [168] may filtration (GFR), tubular secretion and tubular re-

    further enhance the interindividual variability in drug absorption. At birth, these renal clearance mecha-

    elimination characteristics observed in infants. Poly- nisms are incompletely developed and renal elimina-

    morphisms in drug metabolism and the potential for tion capacity of the newborn is significantly compro-

    enzyme induction and/or inhibition complicate any mised [169171]. During late gestation and early

  • 8/2/2019 New Born Pharmacokinetics

    13/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 679

    postnatal development profound anatomical and average and a slower pattern of GFR development

    functional changes in the kidney greatly enhance during the first 12 weeks postpartum as compared

    renal elimination efficiency in the first few months of with the full-term infant [4,183,196]. With comple-

    life [172,173]. Renal functions demonstrate a rapid tion of nephrogenesis and maturation of glomerular

    maturation and generally reach adult levels before 1 function, enhancements in GFR in the preterm infantyear of age [174176]. Maturation of glomerular will proceed at the same rate as full-term infants

    filtration and renal tubular functions proceed at [183]. However, even by 5 weeks of age the absolute

    different rates and patterns resulting in marked value for GFR remains lower in preterm infants

    interindividual variability in renal elimination ef- [183]. This functional delay in GFR in preterm

    ficiency. infants is an important consideration in the estima-

    The anatomical and functional development of the tion of an infants capacity for renal elimination.

    kidney continues throughout gestation into the early Interestingly, Fig. 4 illustrates infant GFR, on an

    postnatal period. Nephrons increase in number until ml/ min/ kg basis, is roughly comparable to the adult

    nephrogenesis is completed at 36 weeks of gestation [185]. This implies adult renal clearance values

    [172,173,177,178]. Prior to 36 weeks gestation, then, normalized to a body weight may reasonably predict

    changes in renal function principally correlate with infant renal clearances on a body weight basis.

    increases in the number of nephrons [179181].

    Incomplete nephrogenesis in the pre-term newborn 4.2.2. Renal tubular function

    will compromise glomerular and tubular function At birth, the renal tubules exhibit significant

    [182]. Functional maturation and growth processes anatomic and functional immaturity [197]. Incom-

    explain the changes in renal elimination capacity in plete anatomical development of renal tubules com-

    the full-term infant [98,172]. In general, postnatal promises both passive reabsorption [188,198] and

    functional maturation of the kidney is associated active secretion and reabsorption processes [199

    with enhancements in renal blood flow, improve- 201]. In addition to limited tubular size and func-

    ments in glomerular filtration efficiency and the tional maturity, poor peritubular blood flow, reduced

    growth and maturation of renal tubules and tubular urine concentrating ability, and lower urinary pH

    processes [98]. values further compromise renal tubule function in

    the newborn [202]. In general, renal tubular growth4.2.1. Glomerular filtration processes, maturation of renal tubular transport sys-

    During the fetal stages, GFR capacity is signifi- tems, and redistribution of blood flow to the secret-

    cantly reduced [172]. Parturition triggers enhance- ory areas of the kidney account for the enhancements

    ments in both cardiac output and renal blood flow

    and a dramatic decrease in renal vascular resistance

    and a redistribution of blood flow within the kidney

    [172,183185]. These hemodynamic changes cause a

    rapid increase in GFR during the early postnatal

    period [4,171,186190]. At birth, GFR, normalized

    to body surface area, in the full-term infant is 10152

    ml/ min/ m [169,171], but increases to 2030 ml /2min/ m within the first 2 weeks of life [171,191]. By

    6 months of age, infant GFR, normalized to body

    surface area has approached adult levels (73 ml/2

    min/m ) [192]. Rapid improvements in GFR result

    in rapid enhancements in the renal clearance ofFig. 4. General pattern of postnatal development of the renalcompounds principally eliminated by GFR.clearance pathways in newborns and infants. Developmental

    Postnatal improvements in GFR correlate withchanges in glomerular filtration rate (GFR), renal tubular secretion

    gestational age rather than postnatal age [4,193 (TS) and renal blood flow (Q ). Data adapted from [98] and areR

    195]. Premature infants exhibit lower GFR values on expressed as fraction of adult clearance values (ml/min/kg).

  • 8/2/2019 New Born Pharmacokinetics

    14/20

    680 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    in renal tubular function during postnatal develop- tional immaturity of absorption, distribution, metabo-

    ment [171,187,203]. As the infant develops, matura- lism and/or excretion processes contribute to the

    tion of renal tubular function generally exhibits a disparate responses observed between newborns,

    more protracted time course than GFR. This infants and adults. Premature infants present a fur-

    produces a functional glomerulotubular imbalance ther complication as the anatomical and functionaluntil renal tubule maturation is completed by 1 year immaturity of the organs and other biochemical and

    of age [204,205]. physiological processes involved in drug phar-

    Numerous protein carrier systems mediate active macokinetics is further exacerbated. An assessment

    renal excretion and reabsorption. Their postnatal of the therapeutic efficacy or toxicant susceptibility

    development at the renal tubular epithelium and their of a newborn to an exposure will require a careful

    impact on renal elimination efficiency in the new- consideration of the developmental aspects of phar-

    born and infant remains largely unknown. Func- macokinetic processes. In general, the combined

    tionally, the kidney exhibits a reduced capacity to effects of age-related changes in each phar-

    excrete weak organic acids like penicillins, sulfon- macokinetic process on plasma levels of a compound

    amides, and cephalosporins [200,201]. Newborn are poorly understood. Clinical studies encompassing

    kidneys excrete p-aminohippurate (PAH), a substrate newborns and infants within narrow postnatal age

    for the organic anion transporters, at 2030% adult groups are needed to enhance our understanding of

    levels [187], and adult excretion levels are ap- the pharmacokinetic and clinical consequences of

    proached by 78 months of age [176]. Premature postnatal maturation of absorption, distribution, me-

    and full-term infants excrete furosemide, a PAH tabolism and excretion processes. Such information

    transport pathway substrate, slowly with plasma half- will help to establish more effective guidelines to

    lives of 19.9 and 7.7 h, respectively, as compared predict an exposure outcome in a newborn or young

    with 0.5 h in the adult [206,207]. In utero or infant infant and to ensure safe exposures to therapeutic or

    exposure to certain agents may induce or inhibit inadvertent compounds.

    renal tubular transport functions [174]. Transport

    induction or inhibition may compound the variability

    observed in renal clearance values in newborns and References

    young infants. The low urinary pH values relative tothe adult may influence the reabsorption of weak [1] E. Zecca, P. Papacci, L. Maggio, F. Gallini, S. Elia, G. De

    Rosa, C. Romagnoli, Cardiac adverse effects of early dexa-organic acids and bases, and differences in renalmethasone treatment in preterm infants: a randomized clini-drug elimination may reflect a discrepancy in urinarycal trial, J. Clin. Pharmacol. 41 (2001) 10751081.

    pH values [166].[2] L. Salvaneschi, C. Perotti, M. Zecca, S. Bernuzzi, G.

    The anatomical and functional immaturity of the Viarengo, G. Giorgiani, C. Del Fante, P. Bergamaschi, R.newborn kidney leads to reduced renal clearances of Maccario, A. Pession, F. Locatelli, Extracorporeal photo-

    chemotherapy for treatment of acute and chronic GVHD incompounds during the early postnatal period. Differ-childhood, Transfusion 41 (2001) 12991305.ences in the rate of development of glomerular

    [3] I. Choonara, S. Conroy, Unlicensed and off-label drug use infiltration and tubular function (i.e., the

    children: implications for safety, Drug Saf. 25 (2002) 15.glomerulotubular imbalance) and the potential for the [4] J.N. van den Anker, Pharmacokinetics and renal function in

    induction or inhibition of renal glomerular and tubule preterm infants, Acta Paediatr. 85 (1996) 13931399.[5] C.L. Berseth, Effect of early feeding on maturation of thetransport function [208] may have variable andpreterm infants small intestine, J. Pediatr. 120 (1992) 947complex effects on the renal elimination.953.

    [6] R.C. Dumont, C.D. Rudolph, Development of gastrointesti-

    nal motility in the infant and child, Gastroenterol. Clin.

    5. Conclusions North Am. 23 (1994) 655671.[7] J.B. Besunder, M.D. Reed, J.L. Blumer, Principles of drug

    biodisposition in the neonate. A critical evaluation of thePostnatal maturation of pharmacokinetic processespharmacokineticpharmacodynamic interface (Part I), Clin.

    has significant implications with respect to systemicPharmacokinet. 14 (1988) 189216.

    exposure levels and the safety and/or efficacy of a [8] G. Heimann, Enteral absorption and bioavailability in chil-compound in the newborn and young infant. Func- dren in relation to age, Eur. J. Clin. Pharmacol. 18 (1980)

    4350.

  • 8/2/2019 New Born Pharmacokinetics

    15/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 681

    [9] D.W. Ebers, D.I. Smith, G.E. Gibbs, Gastric acidity on the [29] P.J. Milla, Intestinal motility during ontogeny and intestinal

    first day of life, Pediatrics 18 (1956) 800805. pseudo-obstruction in children, Pediatr. Clin. North Am. 43[10] G.B. Avery, J.G. Randolph, T. Weaver, Gastric acidity in the (1996) 511532.

    first day of life, Pediatrics 37 (1966) 10051007. [30] I. Blumenthal, R.S. Pildes, Effect of posture on the pattern of[11] M. Agunod, N. Yamaguchi, R. Lopez, A.L. Luhby, G.B. stomach emptying in the newborn, Pediatrics 63 (1979)

    Glass, Correlative study of hydrochloric acid, pepsin, and 532536.intrinsic factor secretion in newborns and infants, Am. J. [31] D.R. Krishna, U. Klotz, Extrahepatic metabolism of drugs inDig. Dis. 14 (1969) 400414. humans, Clin. Pharmacokinet. 26 (1994) 144160.

    [12] A.R. Euler, W.J. Byrne, L.M. Cousins, M.E. Ament, J.H. [32] G.L. Simon, S.L. Gorbach, Intestinal flora in health andWalsh, Increased serum gastrin concentrations and gastric disease, Gastroenterology 86 (1984) 174193.acid hyposecretion in the immediate newborn period, Gas-

    [33] M.M. Gronlund, S. Salminen, H. Mykkanen, P. Kero, O.P.troenterology 72 (1977) 12711273.

    Lehtonen, Development of intestinal bacterial enzymes in[13] A.F. Hess, The gastric secretion of infants at birth, Am. J.

    infantsrelationship to mode of delivery and type of feed-Dis. Child. 6 (1913) 264276.

    ing, Apmis 107 (1999) 655660.[14] R.A. Miller, Observations on the gastric acidity during the

    [34] C.T. Huang, J.T. Rodriguez, W.E. Woodward, B.L. Nichols,first month of life, Arch. Dis. Child. 16 (1941) 2230.

    Comparison of patterns of fecal bile acid and neutral sterol[15] M.D. Reed, J.B. Besunder, Developmental pharmacology:

    between children and adults, Am. J. Clin. Nutr. 29 (1976)ontogenic basis of drug disposition, Pediatr. Clin. North Am.

    11961203.36 (1989) 10531074.

    [35] R.S. Obach, Q.Y. Zhang, D. Dunbar, L.S. Kaminsky, Meta-[16] J.S. Deren, Development of structure and function in the bolic characterization of the major human small intestinalfetal and newborn stomach, Am. J. Clin. Nutr. 24 (1971)

    cytochrome p450s, Drug Metab. Dispos. 29 (2001) 347352.144159.

    [36] J.C. Kolars, P. Schmiedlin-Ren, J.D. Schuetz, C. Fang, P.B.[17] N.N. Huang, R.H. High, Comparison of serum levels follow-

    Watkins, Identification of rifampin-inducible P450IIIA4ing the administration of oral and parenteral preparations of

    (CYP3A4) in human small bowel enterocytes, J. Clin. Invest.penicillin to infants and children of various age groups, J.

    90 (1992) 18711878.Pediatr. 42 (1953) 657668.

    [37] Q.Y. Zhang, D. Dunbar, A. Ostrowska, S. Zeisloft, J. Yang,[18] J. Silverio, J.W. Poole, Serum concentrations of ampicillin in

    L.S. Kaminsky, Characterization of human small intestinalnewborn infants after oral administration, Pediatrics 51

    cytochromes P-450, Drug Metab. Dispos. 27 (1999) 804(1973) 578580.

    809.[19] P.L. Morselli, R. Franco-Morselli, L. Bossi, Clinical phar-

    [38] T. Prueksaritanont, L.M. Gorham, J.H. Hochman, L.O. Tran,macokinetics in newborns and infants. Age-related differ-K.P. Vyas, Comparative studies of drug-metabolizing en-ences and therapeutic implications, Clin. Pharmacokinet. 5zymes in dog, monkey, and human small intestines, and in(1980) 485527.Caco-2 cells, Drug Metab. Dispos. 24 (1996) 634642.[20] A. Wallin, B. Jalling, L.O. Boreus, Plasma concentrations of

    [39] S. Madani, M.F. Paine, L. Lewis, K.E. Thummel, D.D. Shen,phenobarbital in the neonate during prophylaxis for neonatalComparison of CYP2D6 content and metoprolol oxidationhyperbilirubinemia, J. Pediatr. 85 (1974) 392397.between microsomes isolated from human livers and small[21] P. McClean, L.T. Weaver, Ontogeny of human pancreaticintestines, Pharm. Res. 16 (1999) 11991205.exocrine function, Arch. Dis. Child. 68 (1993) 6265.

    [40] T.N. Johnson, M.S. Tanner, C.J. Taylor, G.T. Tucker,[22] E. Lebenthal, P.C. Lee, Development of functional responsesEnterocytic CYP3A4 in a paediatric population: develop-in human exocrine pancreas, Pediatrics 66 (1980) 556560.mental changes and the effect of coeliac disease and cystic[23] L. Barbara, R. Lazzari, A. Roda, R. Aldini, D. Festi, C.fibrosis, Br. J. Clin. Pharmacol. 51 (2001) 451460.Sama, A.M. Morselli, A. Collina, F. Bazzoli, G. Mazzella, E.

    [41] H.Y. Yang, Q.P. Lee, A.E. Rettie, M.R. Juchau, FunctionalRoda, Serum bile acids in newborns and children, Pediatr.cytochrome P4503A isoforms in human embryonic tissues:Res. 14 (1980) 12221225.expression during organogenesis, Mol. Pharmacol. 46 (1994)[24] G.M. Murphy, E. Signer, Bile acid metabolism in infants and922928.children, Gut 15 (1974) 151163.

    [42] H. Hoensch, I. Morgenstern, G. Petereit, M. Siepmann, W.H.[25] J.B. Watkins, D. Ingall, P. Szczepanik, P.D. Klein, R. Lester, Peters, H.M. Roelofs, W. Kirch, Influence of clinical factors,Bile-salt metabolism in the newborn. Measurement of pooldiet, and drugs on the human upper gastrointestinal gluta-size and synthesis by stable isotope technic, New Engl. J.thione system, Gut 50 (2002) 235240.Med. 288 (1973) 431434.

    [43] W.C. de Bruin, M.J. Wagenmans, W.H. Peters, Expression of[26] G. Boehm, M. Borte, H. Muller, G. Moro, I. Minoli,glutathione S-transferase alpha, P1-1 and T1-1 in the humanActivities of trypsin and lipase in duodenal aspirates ofgastrointestinal tract, Jpn. J. Cancer Res. 91 (2000) 310316.preterm infants: influence of dietary protein and fat com-

    [44] M. Cappiello, L. Giuliani, G.M. Pacifici, Differential dis-position, Am. J. Clin. Nutr. 61 (1995) 524527.

    tribution of phenol and catechol sulphotransferases in human[27] C.L. Berseth, Gestational evolution of small intestine motili-liver and intestinal mucosa, Pharmacology 40 (1990) 6976.ty in preterm and term infants, J. Pediatr. 115 (1989) 646

    651. [45] P.J. Czernik, J.M. Little, G.W. Barone, J.P. Raufman, A.

    [28] M. Gupta, Y.W. Brans, Gastric retention in neonates, Pediat- Radominska-Pandya, Glucuronidation of estrogens and reti-

    rics 62 (1978) 2629. noic acid and expression of UDP-glucuronosyltransferase

  • 8/2/2019 New Born Pharmacokinetics

    16/20

    682 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    2B7 in human intestinal mucosa, Drug Metab. Dispos. 28 tion of midazolam involves both gastrointestinal and hepatic

    (2000) 12101216. CYP3A-mediated metabolism, Clin. Pharmacol. Ther. 59

    (1996) 491502.[46] M.B. Fisher, M. Vandenbranden, K. Findlay, B. Burchell,

    K.E. Thummel, S.D. Hall, S.A. Wrighton, Tissue distribution [62] A. Tsuji, I. Tamai, Carrier-mediated intestinal transport of

    and interindividual variation in human UDP- drugs, Pharm. Res. 13 (1996) 963977.

    glucuronosyltransferase activity: relationship between [63] S.D. Hall, K.E. Thummel, P.B. Watkins, K.S. Lown, L.Z.UGT1A1 promoter genotype and variability in a liver bank, Benet, M.F. Paine, R.R. Mayo, D.K. Turgeon, D.G. Bailey,Pharmacogenetics 10 (2000) 727739. R.J. Fontana, S.A. Wrighton, Molecular and physical mecha-

    [47] G.M. Pacifici, C. Bencini, A. Rane, Acetyltransferase in nisms of first-pass extraction, Drug Metab. Dispos. 27

    humans: development and tissue distribution, Pharmacology (1999) 161166.

    32 (1986) 283291. [64] L.Z. Benet, C.L. Cummins, The drug efflux-metabolism

    [48] S.J. Yaffe, M.R. Juchau, Perinatal pharmacology, Annu. Rev. alliance: biochemical aspects, Adv. Drug Deliv. Rev. 50

    Pharmacol. 14 (1974) 219238. (Suppl. 1) (2001) S3S11.

    [49] http://www.med.rug.nl/mdl/ (2002). [65] B. Friis-Hansen, Body water compartments in children:

    changes during growth and related changes in body com-[50] http://lab.digibench.net/transporter/ (2002).

    position, Pediatrics 28 (1961) 169181.[51] M.M. Doherty, W.N. Charman, The mucosa of the small

    [66] Geigy Scientific Tables, 8th Edition, CIBA-GEIGY Limitedintestine: how clinically relevant as an organ of drug

    Basle, Switzerland, 1981.metabolism?, Clin. Pharmacokinet. 41 (2002) 235253.

    [67] B. Friis-Hansen, Body composition during growth. In vivo[52] F. Martel, M.J. Martins, C. Calhau, C. Hipolito-Reis, I. measurements and biochemical data correlated to differentialAzevedo, Postnatal development of organic cation transport

    anatomical growth, Pediatrics 47 (Suppl. 2) (1971) 264.in the rat liver, Pharmacol. Res. 37 (1998) 131136.

    [68] http://www.cdc.gov/nchs/about/major/nhanes/[53] A. Dutt, T.S. Priebe, L.D. Teeter, M.T. Kuo, J.A. Nelson,growthcharts/charts.htm[Set%201 (2002).Postnatal development of organic cation transport and mdr

    gene expression in mouse kidney, J. Pharmacol. Exp. Ther. [69] C.K. Taketomo, J.H. Hodding, D.M. Kraus, Pediatric Dosage

    261 (1992) 12221230. Handbook: Including Neonatal Dosing, Drug Administration

    and Extemporaneous Preparations, 8th Edition, Lexi-Comp[54] T. Kojima, M. Nishimura, T. Yajima, T. Kuwata, Y. Suzuki,

    Inc. and American Pharmaceutical Association, Hudson, OH,T. Goda, S. Takase, E. Harada, Developmental changes in2001.the regional Na1/glucose transporter mRNA along the

    [70] B.M. Assael, Pharmacokinetics and drug distribution duringsmall intestine of suckling rats, Comp. Biochem. Physiol. B

    postnatal development, Pharmacol. Ther. 18 (1982) 159Biochem. Mol. Biol. 122 (1999) 8995.

    197.[55] Y. Matsuoka, M. Okazaki, Y. Kitamura, T. Taniguchi,

    [71] E.M. Cornford, W.M. Pardridge, L.D. Braun, W.H. Old-Developmental expression of P-glycoprotein (multidrug re-

    endorf, Increased bloodbrain barrier transport of protein-sistance gene product) in the rat brain, J. Neurobiol. 39bound anticonvulsant drugs in the newborn, J. Cereb. Blood(1999) 383392.

    Flow Metab. 3 (1983) 280286.[56] A. Pavlova, H. Sakurai, B. Leclercq, D.R. Beier, A.S. Yu,

    [72] J.R. Koup, B.A. Hart, Relationship between plasma andS.K. Nigam, Developmentally regulated expression of or-

    whole blood theophylline concentration in neonates, J.ganic ion transporters NKT (OAT1), OCT1, NLT (OAT2)

    Pediatr. 94 (1979) 320321.and Roct, Am. J. Physiol. Renal Physiol. 278 (2000) F635

    F643. [73] I. Elbourne, E.R. Lumbers, K.J. Hill, The secretion of

    organic acids and bases by the ovine fetal kidney, Exp.[57] C. van Kalken, G. Giaccone, P. van der Valk, C.M. Kuiper,

    Physiol. 75 (1990) 211221.M.M. Hadisaputro, S.A. Bosma, R.J. Scheper, C.J. Meijer,

    H.M. Pinedo, Multidrug resistance gene (P-glycoprotein) [74] W.F. Balistreri, Immaturity of hepatic excretory function and

    expression in the human fetus, Am. J. Pathol. 141 (1992) the ontogeny of bile acid metabolism, J. Pediatr. Gastroen-

    10631072. terol. Nutr. 2 (1983) S207S214.

    [58] U. Schumacher, K. Mollgard, The multidrug-resistance P- [75] C. Dubuisson, D. Cresteil, M. Desrochers, D. Decimo, M.

    glycoprotein (Pgp, MDR1) is an early marker of bloodbrain Hadchouel, E. Jacquemin, Ontogenic expression of thebarrier development in the microvessels of the developing Na(1)-independent organic anion transporting polypeptide

    human brain, Histochem. Cell Biol. 108 (1997) 179182. (oatp) in rat liver and kidney, J. Hepatol. 25 (1996) 932

    940.[59] C.Y. Wu, L.Z. Benet, M.F. Hebert, S.K. Gupta, M. Rowland,

    D.Y. Gomez, V.J. Wacher, Differentiation of absorption and [76] E.M. Widdowson, Changes in body proportions and com-

    first-pass gut and hepatic metabolism in humans: studies with position during growth, in: J.A. Davies, J. Dobbing (Eds.),

    cyclosporine, Clin. Pharmacol. Ther. 58 (1995) 492497. Scientific Foundations of Pediatrics, Heinemann Medical

    Books Ltd, London, 1974, pp. 153163.[60] M.F. Paine, D.D. Shen, K.L. Kunze, J.D. Perkins, C.L.

    Marsh, J.P. McVicar, D.M. Barr, B.S. Gillies, K.E. Thummel, [77] S. Curry, P. Brick, N.P. Franks, Fatty acid binding to human

    First-pass metabolism of midazolam by the human intestine, serum albumin: new insights from crystallographic studies,

    Clin. Pharmacol. Ther. 60 (1996) 1424. Biochim. Biophys. Acta 1441 (1999) 131140.

    [61] K.E. Thummel, D. OShea, M.F. Paine, D.D. Shen, K.L. [78] T. Fournier, N.N. Medjoubi, D. Porquet, Alpha-1-acid

    Kunze, J.D. Perkins, G.R. Wilkinson, Oral first-pass elimina- glycoprotein, Biochim. Biophys. Acta 1482 (2000) 157171.

    http://www.med.rug.nl/mdl/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.cdc.gov/nchs/about/major/nhanes/growthcharts/charts.htm#Set%201http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.lab.digibench.net/transporter/http://www.med.rug.nl/mdl/http://www.med.rug.nl/mdl/http://www.med.rug.nl/mdl/http://www.med.rug.nl/mdl/http://www.med.rug.nl/mdl/http://www.med.rug.nl/mdl/http://www.med.rug.nl/mdl/
  • 8/2/2019 New Born Pharmacokinetics

    17/20

    J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686 683

    [79] J.M. Kremer, J. Wilting, L.H. Janssen, Drug binding to metabolizing enzymes: phase I oxidative enzymes, J. Phar-

    macol. Exp. Ther. 300 (2002) 355360.human alpha-1-acid glycoprotein in health and disease,[97] S.N. de Wildt, G.L. Kearns, J.S. Leeder, J.N. van den Anker,Pharmacol. Rev. 40 (1988) 147.

    Glucuronidation in humans. Pharmacogenetic and develop-[80] P.A. Routledge, Clinical relevance of alpha 1 acid glycopro-mental aspects, Clin. Pharmacokinet. 36 (1999) 439452.tein in health and disease, Prog. Clin. Biol. Res. 300 (1989)

    [98] W.L. Hayton, Maturation and growth of renal function:185198.dosing renally cleared drugs in children, AAPS Pharmsci. 2[81] M. Ehrnebo, S. Agurell, B. Jalling, L.O. Boreus, Age(2000) article 3.differences in drug binding by plasma proteins: studies on

    [99] K.S. Pang, M. Rowland, Hepatic clearance of drugs. I.human foetuses, neonates and adults, Eur. J. Clin. Phar-Theoretical considerations of a well-stirred model and amacol. 3 (1971) 189193.parallel tube model. Influence of hepatic blood flow, plasma[82] L. Herngren, M. Ehrnebo, L.O. Boreus, Drug binding toand blood cell binding, and the hepatocellular enzymaticplasma proteins during human pregnancy and in the perinatalactivity on hepatic drug clearance, J. Pharmacokinet. Bio-period. Studies on cloxacillin and alprenolol, Dev. Phar-pharm. 5 (1977) 625653.macol. Ther. 6 (1983) 110124.

    [100] M. Yamazaki, H. Suzuki, Y. Sugiyama, Recent advances in[83] G.M. Pacifici, A. Viani, G. Taddeucci-Brunelli, Serum

    carrier-mediated hepatic uptake and biliary excretion ofprotein binding of furosemide in newborn infants and

    xenobiotics, Pharm. Res. 13 (1996) 497513.children, Dev. Pharmacol. Ther. 10 (1987) 413421.

    [101] S.Z. Cagen, J.E. Gibson, Characteristics of hepatic excret-[84] H. Echizen, M. Nakura, T. Saotome, S. Minoura, T. Ishizaki,

    ory function during development, J. Pharmacol. Exp. Ther.Plasma protein binding of disopyramide in pregnant and

    210 (1979) 1521.postpartum women, and in neonates and their mothers, Br. J.[102] E. Fischer, A. Barth, F. Varga, W. Klinger, Age dependence

    Clin. Pharmacol. 29 (1990) 423430.of hepatic transport in control and phenobarbital-pretreated

    [85] F. Kanakoudi, V. Drossou, V. Tzimouli, E. Diamanti, T.rats, Life Sci. 24 (1979) 557562.

    Konstantinidis, A. Germenis, G. Kremenopoulos, Serum[103] R.T. Williams, Detoxification Mechanisms, Wiley and Sons,

    concentrations of 10 acute-phase proteins in healthy term andNew York, 1959.

    preterm infants from birth to age 6 months, Clin. Chem. 41[104] D. Ratanasavanh, P. Beaune, F. Morel, J.P. Flinois, F.P.

    (1995) 605608.Guengerich, A. Guillouzo, Intralobular distribution and

    [86] P.J. McNamara, J. Alcorn, Protein binding predictions inquantitation of cytochrome P-450 enzymes in human liver

    infants, AAPS Pharm. Sci. 4 (2002) article 4.as a function of age, Hepatology 13 (1991) 11421151.

    [87] H. Nau, W. Luck, W. Kuhnz, Decreased serum protein[105] http:/ /drnelson.utmem.edu/ CytochromeP450.html, David

    binding of diazepam and its major metabolite in the neonateNelsons Cytochrome P450 Homepage (2002).

    during the first postnatal week relate to increased free fatty[106] T. Shimada, H. Yamazaki, M. Mimura, Y. Inui, F.P.

    acid levels, Br. J. Clin. Pharmacol. 17 (1984) 9298.Guengerich, Interindividual variations in human liver cyto-

    [88] H. Kurz, H. Michels, H.H. Stickel, Differences in the binding chrome P-450 enzymes involved in the oxidation of drugs,of drugs to plasma proteins from newborn and adult man. II, carcinogens and toxic chemicals: studies with liver micro-Eur. J. Clin. Pharmacol. 11 (1977) 469472. somes of 30 Japanese and 30 Caucasians, J. Pharmacol.

    [89] A.H. Bardy, V.K. Hiilesmaa, K. Teramo, P.J. Neuvonen, Exp. Ther. 270 (1994) 414423.Protein binding of antiepileptic drugs during pregnancy, [107] T. Shimada, H. Yamazaki, M. Mimura, N. Wakamiya, Y.F.labor and puerperium, Ther. Drug Monit. 12 (1990) 4046. Ueng, F.P. Guengerich, Y. Inui, Characterization of micro-

    [90] A. Rane, P.K. Lunde, B. Jalling, S.J. Yaffe, F. Sjoqvist, somal cytochrome P450 enzymes involved in the oxidationPlasma protein binding of diphenylhydantoin in normal and of xenobiotic chemicals in human fetal liver and adulthyperbilirubinemic infants, J. Pediatr. 78 (1971) 877882. lungs, Drug Metab. Dispos. 24 (1996) 515522.

    [91] J. Alcorn, P.J. McNamara, The ontogeny of hepatic and renal [108] L.Z. Benet, D.L. Kroetz, L.B. Sheiner, Pharmacokinetics.systemic clearance pathways in infants: A review (Part I), The dynamics of drug absorption, disposition and elimina-Clin. Pharmacokinet. 41 (2002) 959998. tion, in: J.G. Hardman, L.E. Limbird (Eds.), Goodman and

    [92] J. Hakkola, E. Tanaka, O. Pelkonen, Developmental expres- Gilmans the Pharmacological Basis of Therapeutics, 9th

    sion of cytochrome P450 enzymes in human liver, Phar- Edition, McGraw-Hill, New York, 1995, pp. 327.macol. Toxicol. 82 (1998) 209217. [109] J.T. Gilman, Therapeutic drug monitoring in the neonate[93] J.A. Ring, H. Ghabrial, M.S. Ching, R.A. Smallwood, D.J. and paediatric age group. Problems and clinical phar-

    Morgan, Fetal hepatic drug elimination, Pharmacol. Ther. 84 macokinetic implications, Clin. Pharmacokinet. 19 (1990)(1999) 429445. 110.

    [94] P.J. Gow, H. Ghabrial, R.A. Smallwood, D.J. Morgan, M.S. [110] G.L. Kearns, M.D. Reed, Clinical pharmacokinetics inChing, Neonatal hepatic drug elimination, Pharmacol. Tox- infants and children. A reappraisal, Clin. Pharmacokinet. 17icol. 88 (2001) 315. (1989) 2967.

    [95] D.G. McCarver, R.N. Hines, The ontogeny of human drug- [111] D. Lacroix, M. Sonnier, A. Moncion, G. Cheron, T.

    metabolizing enzymes: phase II conjugation enzymes and Cresteil, Expression of CYP3A in the human liverevi-

    regulatory mechanisms, J. Pharmacol. Exp. Ther. 300 (2002) dence that the shift between CYP3A7 and CYP3A4 occurs

    361366. immediately after birth, Eur. J. Biochem. 247 (1997) 625

    634.[96] R.N. Hines, D.G. McCarver, The ontogeny of human drug-

    http://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.htmlhttp://www.drnelson.utmem.edu/CytochromeP450.html
  • 8/2/2019 New Born Pharmacokinetics

    18/20

    684 J. Alcorn, P.J. McNamara / Advanced Drug Delivery Reviews 55 (2003) 667686

    [112] M. Sonnier, T. Cresteil, Delayed ontogenesis of CYP1A2 in Kanakubo, P-450 HFLa, a form of cytochrome P-450

    the human liver, Eur. J. Biochem. 251 (1998) 893898. purified from human fetal livers, is the 16 alpha-hydroxy-[113] J.M. Treluyer, E. Jacqz-Aigrain, F. Alvarez, T. Cresteil, lase of dehydroepiandrosterone 3-sulfate, J. Biol. Chem.

    Expression of CYP2D6 in developing human liver, Eur. J. 262 (1987) 1353413537.Biochem. 202 (1991) 583588. [129] M. Kitada, T. Kamataki, K. Itahashi, T. Rikihisa, Y.

    [114] J.M. Treluyer, G. Gueret, G. Cheron, M. Sonnier, T. Kanakubo, Significance of cytochrome P-450 (P-450 HFLa)Cresteil, Developmental expression of CYP2C and CYP2C- of human fetal livers in the steroid and drug oxidations,dependent activities in the human liver: in-vivo/ in-vitro Biochem. Pharmacol. 36 (1987) 453456.correlation and inducibility, Pharmacogenetics 7 (1997) [130] H. Hashimoto, K. Toide, R. Kitamura, M. Fujita, S.441452. Tagawa, S. Itoh, T. Kamataki, Gene structure of CYP3A4,

    [115] I. Vieira, M. Sonnier, T. Cresteil, Developmental expression an adult-specific form of cytochrome P450 in human livers,of CYP2E1 in the human liver. Hypermethylation control and its transcriptional control, Eur. J. Biochem. 218 (1993)of gene expression during the neonatal period, Eur. J. 585595.Biochem. 238 (1996) 476483.

    [131] S. Ohmori, N. Fujiki, H. Nakasa, H. Nakamura, I. Ishii, K.[116] T. Cresteil, Onset of xenobiotic metabolism in children:

    Itahashi, M. Kitada, Steroid hydroxylation by human fetaltoxicological implications, Food Addit. Contam. 15 (1998)

    CYP3A7 and human NADPH-cytochrome P450 reductase4551.

    coexpressed in insect cells using baculovirus, Res. Com-[117] J.M. Treluyer, G. Cheron, M. Sonnier, T. Cresteil, Cyto-

    mun. Mol. Pathol. Pharmacol. 100 (1998) 1528.chrome P-450 expression in sudden infant death syndrome,

    [132] P. Kuehl, J. Zhang, Y. Lin, J. Lamba, M. Assem, J. Schuetz,Biochem. Pharmacol. 52 (1996) 497504. P.B. Watkins, A. Daly, S.A. Wrighton, S.D. Hall, P. Maurel,[118] T. Cresteil, P. Beaune, P. Kremers, C. Celier, F.P.

    M. Relling, C. Brimer, K. Yasuda, R. Venkataramanan, S.Guengerich, J.P. Leroux, Immunoquantification of epoxide

    Strom, K. Thummel, M.S. Boguski, E. Schuetz, Sequencehydrolase and cytochrome P-450 isozymes in fetal and

    diversity in CYP3A promoters and characterization of theadult human liver microsomes, Eur. J. Biochem. 151 (1985)

    genetic basis of polymorphic CYP3A5 expression, Nat.345350.

    Genet. 27 (2001) 383391.[119] O. Pelkonen, Biotransformation of xenobiotics in the fetus,

    [133] P. Burtin, E. Jacqz-Aigrain, P. Girard, R. Lenclen, J.F.Pharmacol. Ther. 10 (1980) 261281.

    Magny, P. Betremieux, C. Tehiry, L. Desplanques, P.[120] T. Tateishi, H. Nakura, M. Asoh, M. Watanabe, M. Tanaka,

    Mussat, Population pharmacokinetics of midazolam inT. Kumai, S. Takashima, S. Imaoka, Y. Funae, Y. Yabusaki,

    neonates, Clin. Pharmacol. Ther. 56 (1994) 615625.T. Kamataki, S. Kobayashi, A comparison of hepatic

    [134] K. Payne, F.J. Mattheyse, D. Liebenberg, T. Dawes, Thecytochrome P450 protein ex