Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 6379 2023-12-24 02:06:15 |
2 format change Meta information modification 6379 2023-12-25 03:20:20 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhang, W.; Zhang, Q.; Cao, Z.; Zheng, L.; Hu, W. Neonatal Physiologic Changes Affecting Drug Disposition Process. Encyclopedia. Available online: https://encyclopedia.pub/entry/53103 (accessed on 17 May 2024).
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Neonatal Physiologic Changes Affecting Drug Disposition Process. Encyclopedia. Available at: https://encyclopedia.pub/entry/53103. Accessed May 17, 2024.
Zhang, Wei, Qian Zhang, Zhihai Cao, Liang Zheng, Wei Hu. "Neonatal Physiologic Changes Affecting Drug Disposition Process" Encyclopedia, https://encyclopedia.pub/entry/53103 (accessed May 17, 2024).
Zhang, W., Zhang, Q., Cao, Z., Zheng, L., & Hu, W. (2023, December 24). Neonatal Physiologic Changes Affecting Drug Disposition Process. In Encyclopedia. https://encyclopedia.pub/entry/53103
Zhang, Wei, et al. "Neonatal Physiologic Changes Affecting Drug Disposition Process." Encyclopedia. Web. 24 December, 2023.
Neonatal Physiologic Changes Affecting Drug Disposition Process
Edit

Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles between neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. 

modeling and simulation term infants preterm infants

1. Introduction

The lack of pediatric drug research is due to difficulties in recruiting a sufficient number of children for clinical studies, ethical concerns associated with enrolling younger children, low consent rates from neonatal parents, limited blood volume availability, and high costs [1]. Consequently, fewer drugs have been authorized and are available for pediatric clinical therapy as compared to the drugs available for adults.
Children are not simply smaller adults because the difference between them is not only due to body weight or surface area but also due to physiological and biochemical changes according to age, particularly in early life [2][3]. Childhood is characterized by pronounced differences in growth and development [4]. Compared to other pediatric subpopulations, the weight of infants from term birth to two years of age increases dramatically by approximately three times, and premature neonates can triple their weight several weeks after receiving postnatal care [5]. Growth rates are most prominent during the last trimester of pregnancy and the first trimester after delivery and are much higher than the pubertal growth spurt [6]. Additionally, maturational physiological changes in structural and functional development include the development of organ system functions, cardiac output (CO), organ perfusion, permeability, glomerular filtration rate (GFR), and the ontogeny of metabolic enzymes and transport [7]. Physiological factors in specific neonatal subpopulations differ significantly from those in general adult and pediatric populations, resulting in alterations to the main pharmacokinetic (PK) processes, including absorption, distribution, metabolism, and excretion (ADME). For example, increased gastric pH and emptying time and decreased gastrointestinal size and transport time are known to reduce the absorption rate of biopharmaceutical classification system class I and II compounds in preterm neonates [8][9][10]. The reported lower plasma protein concentration, higher body water content, and lower blood volume affect drug distribution in neonates [5]. Studies also have shown age-related changes in the activity and expression of uridine-5-diphosphate glucuronosyltransferases (UGTs) and cytochrome p450 (CYP450) enzymes, resulting in reduced drug clearance [11][12]. Neonatal renal clearance systems, such as GFR, glomerular permeability, and reabsorption, are immature and change rapidly throughout the first few months of life, reducing drug excretion [13]. These growth and development processes are not linear and display interindividual variation [14]. However, there are still many knowledge gaps regarding the ontogeny of the organs or tissues involved in ADME processes, the effects of diseases on physiological maturation, and the influence of developmental factors on drug disposition and pharmacological effects. Therefore, one of the greatest difficulties in pediatric drug therapy is constantly exploring and adjusting the proper, safe, and effective dose for neonates using evidence-based information rather than empirical treatment.
Traditionally, allometric principles based on age, body weight, and body surface area have been widely used to extrapolate suitable doses for children from adult data. Using the power function, allometric scaling can reflect the relation between PK parameters, such as clearance, volume of distribution, half-life, and size for different ages, from preterm neonates to adults [15][16][17][18][19][20]. Previously, the allometric exponent used for all age groups was fixed as 0.75 [21][22]. At present, this exponent is challenged for younger children (<2 years of age), and the emerging age-dependent exponent model improved predictive performance by applying 1.2 as the allometric exponent for preterm, 1.1 for term neonates (≤3 months of age), and 1.0 for children > 3 months to 2 years [15]. However, allometry scaling cannot accurately describe the developmental changes in the drug clearance system, particularly drug-metabolizing enzymes, as well as the relation between the concentration effect and response, which may result in misestimation of drug kinetics, imprecise dosing of drugs, drug-induced toxicity, or reduced efficacy in younger children [23].

2. Drug Absorption

Absorption describes the concentration–time pattern when administered through nonvascular routes, such as oral, intramuscular, percutaneous, intranasal, and rectal routes, and is usually expressed as bioavailability and absorption rate. Suppose drugs are administered intramuscularly. In this case, the absorption rate of neonates is challenging to estimate because skeletal muscle blood flow and muscular contraction are reported to be reduced, and the body water content and capillary density in skeletal muscles are reported to be much higher in neonates [24][25]. The common drugs administrated intramuscularly for neonates are vitamin K formulations and vaccines. The extent of transdermal formulation absorption is inversely correlated to the thickness of epidermis and directly related to the degree of skin hydration and skin surface area to body weight ratio [25]. The epidermal development is dependent on gestational age (GA) and is complete at the 34th gestational week (GW) [26]. The full-term neonates have intact skin barrier function while preterm infants lack the vernix caseosa and are more sensitive to percutaneous toxicity [26]. Hence, percutaneous administration of drugs must be conservative in the first few weeks of life. Rectal administration is a good alternative in emergency situations when neonates are unconscious, uncooperative, or vomiting. Due to the fact that rectal administration is a less invasive route, it is well accepted in infants. However, the high variability of drug exposure in neonates limits the use of rectal administration, although this route can avoid first-pass metabolism. Previous study has showed that the absorption rate and extent of rectal acetaminophen are lower than oral administration in preterm and term neonates [27]. Most full-term newborns have entered the alveolar development stage after birth, whereas highly preterm infants are at the bronchiole and alveolar epithelial development stage and late preterm infants are at the saccular phase of lung development, so the inhalation routine requires caution in premature neonates [28][29]. The rate and extent of oral drug absorption in neonates are affected by a variety of physiological factors, including gastric pH and emptying time, characteristics of gastric juices, small and large intestinal transit time, gastric and intestinal volume, bile production, intestinal membrane transporters, and drug-metabolizing enzymes [30].
Oral administration is the preferred route for pediatric patients, and liquid dosage forms are preferred in neonates because they are easy to administer. The primary sites of oral drug absorption in newborns are the stomach, small intestine, and colon [31].
The structural and functional maturity of the neonatal gastrointestinal tract varies, mainly depending on the term or preterm birth. Drug absorption in the gut is mainly affected by the gastric pH and gastric emptying time. The gastric pH of the neonate drops from approximately 7 to approximately 2 after birth, then rises to above 4 and finally declines back to approximately 2 in two years [32]. Moreover, the gastric pH in preterm neonates is relatively higher than term infants due to lower levels of basal acid and gastric secretions [13]. A relatively high gastric pH in the body may increase absorption of weakly alkaline drugs and reduce absorption of weakly acidic drugs. Gastric motility is low in highly preterm newborns and approaches full-term newborns after 32 weeks of gestation, whereas gastric emptying in neonates is slower than that in older children [33]. Anatomical differentiation of the human gut reaches neonatal levels by 20 weeks of gestation, and functional maturation, such as digestive enzyme secretion or closure of tight junctions, develops at different rates and generally occurs after the GW of 32–34 [34][35]. A previous study showed that small and large intestinal lengths increase from fetal to maturation age [36]. Low production of bile acids and bile salts in the intestinal lumen at birth may affect enterohepatic bile circulation; however, passive reuptake and active transport of bile are also present [37]. Intestinal motility affects intestinal drug absorption by changing the intestinal transit time. An in vitro model suggested that, similar to adults, the intestinal transit time in term neonates was approximately four hours, whereas that in preterm newborns was approximately four-fold longer due to reduced intestinal motility and peristalsis [38]. Specifically, the overall absorption is delayed and incomplete in newborns. The expression of intestinal P-glycoprotein in neonates is low, especially in those born before the 28th GW [39]. Similarly, the protein expression of CYP3A4 is limited after birth and increases from neonates to adults, which may result in an increase in bioavailability in newborns [36][40]. Drug absorption in the gastrointestinal tract is also affected by regional blood flow, especially in critical ill status such as hypoxia. In addition, pancreatic and biliary functions are immature after birth and develop with age. The physiologic changes affecting oral drug absorption in neonates are listed in Table 1.
Table 1. List of physiological changes that affect the oral drug absorption in neonates (preterm and full-term). Fetus: 0 years, neonates: 0–1 months, infants: 1 month–2 years, children: 2–12 years, adolescents 12–16 years, adults: >16 years.

Abbreviations: CYP, cytochrome P450; GW, gestational week.

3. Drug Distribution

Once a drug enters the systemic circulation, it is distributed to various tissues and organs, which is important for the interaction between drugs and targets. Drug distribution involves passive processes, such as protein-binding permeability, and active processes, such as influx and efflux through transporters [37]. Age-related changes in the body composition and function of the cardiovascular system, compound properties, such as lipophilicity, protein binding, and disease status affect drug distribution in vivo [37]. An impressive increase in body weight, length, and surface area has been observed during early life (Table 2). In addition to the changes mentioned above, changes in body composition were predominant in newborns. Water is the major component of cells and tissues, being responsible for about 60–65% of an adult’s weight, and a higher percentage in full-term (80–85%) and preterm infants (90%) [51][52][53]. The contraction of extracellular water with age results in a 5–7% weight loss in term newborns and a higher weight loss of 10–15% in preterm neonates with meager birth weight (<1500 g) at the end of the first week [51]. Combined with the low body fat percentage at birth (10–15%), neonates require larger doses of hydrophilic drugs to achieve similar efficacy because the ratio of water to lipids is higher in newborns [52]. Both these factors are more significant in the preterm infants, resulting in lower drug plasma concentrations in these different compartments using a body weight-based dosing mode [53][54][55]. The amount and type of plasma proteins determine drug distribution and action, because only unbound drugs can be distributed in vivo and exert pharmacological effects by binding to the corresponding receptors [56]. Human serum albumin (HSA) generally binds to acidic exogenous compounds, whereas alpha 1-glycoprotein (AAG) has a high affinity for basic lipophilic compounds. Previous research reported that the concentration of HSA increased until 20 years of age, then started decreasing with age [57]. Preterm neonates have lower serum albumin concentrations (2.36 g/dL in preterm infants born at 23–34 weeks) compared to term babies (3.43 g/dL) for at least the first three months of life [58]. AAG concentrations seem to stay stable at a low level until 260 days of GA and then begin to rise significantly [59]. Therefore, for highly protein-bound drugs, the amount of free drug and the related pharmacological effect in the body appears to increase in preterm and full-term infants. In addition, physiologically elevated endogenous enzymes, such as bilirubin or fatty acids, competitively bind to plasma proteins, resulting in drug displacement and increased unbound drug concentration [60][61]. Diseases, such as cardiogenic shock or patent ductus arteriosus, may affect CO, regional blood flow, or tissue permeability, thereby increasing the volume of distribution and requiring a higher dose of drugs to achieve a sufficient concentration [62][63].
Table 2. List of physiological changes that affect drug distribution in neonates (preterm and full-term). Fetus: 0 years, neonates: 0–1 months, infants: 1 month–2 years, children: 2–12 years, adolescents 12–16 years, adults: >16 years.

Abbreviations: AAG, alpha 1-glycoprotein; GW, gestational week, HAS, human serum albumin.

4. Drug Metabolism

It is generally assumed that the liver is the major site of drug metabolism. Morphogenesis of the liver occurs during the 10-week GA period, the development of smooth endoplasmic reticulum begins at the 10th GW, and hepatocellular hyperplasia and hypertrophy continue until early adulthood [66]. Previous studies found that antipyrine clearance was related to age even after correction for liver weight [67][68]. Therefore, changes in drug clearance in neonates primarily depend on the maturation of transporters, intrinsic activity of liver enzymes, and regional blood flow, rather than solely on liver size [69][70].
Drug-metabolizing enzymes include phase I enzymes (e.g., CYP450 enzymes and non-CYP-mediated iso-enzymes) involving oxidative, reduction, and hydroxylation, and phase II enzymes (such as UGTs) involving glucuronidation, sulfation, methylation, acetylation, or glutathione conjugation reactions [71]. Because of immature drug metabolism, drug toxicity is more significant in newborns and infants than in adults [72]. The main contributors to drug clearance are CYP enzymes, and each CYP isoenzyme has its own expression and activity ontogeny profiles. Overall, the liver microsomal protein content is lower in newborns than in adults and gradually increases with age, reaching the maximum level by approximately 30 years of age [37]. The most abundant CYP450 enzyme in newborns is CYP3A7, which develops in embryonic hepatic tissue as early as a GA of 50–60 days, and its activity gradually decreases with age but is still present in many individuals until the first year of age [73][74][75]. In contrast, CYP3A4, one of the fastest-changing enzyme activities in early life, displays a mirror image pattern: the activity and expression increase in the first week of age, reaching 30% of adult levels at one month [76][77]. Owing to differences in substrate specificity and catalytic activity between CYP3A7 and CYP3A4, the individual metabolic capacity constantly changes during development and maturation. CYP2E1 can be detected in the liver as early as a GA of 93–186 days, and its expression is highly correlated with increased PNA rather than GA [78]. Non-CYP hepatic isoenzymes, including esterase, flavin-containing monooxygenases (FMOs), and alcohol or aldehyde dehydrogenases, play important roles in drug metabolism by mediating oxidative reactions. Carboxylesterases (CES) are important for insecticide detoxication [79]. In a previous study conducted by Pope et al., no significant differences were found in the expression of hepatic CES between the infants group (2–24 months) and adults (20–36 years) [80]. Moreover, hepatic CES activity was ranked as follows: 2 months < 3 months < 20 years < 24 months < 4 months < 36 years < 21 years < 8 months < 34 years < 35 years [80]. However, Yang et al. found that the mRNA and protein expression of hepatic CES was age-dependent, and its activity in adults was approximately four-fold higher than that in children and 10-fold higher than that in children and fetuses, respectively [81]. FMO1, 2, and 3 are active enzymes involved in exogenous metabolism. Similar to the age-related transition from CYP3A7 to CYP3A4 in the liver, the developmental expression of hepatic FMO1 and FMO3 showed the opposite pattern [82]. Using an optimized enzyme immunoassay, the mean aldehyde dehydrogenase content of perinatal infants was found to be approximately 10-fold lower than that in adults [83]. Phase II catalytic enzymes include glucuronosyltransferases, sulfotransferases, glutathione S-transferases, arylamine N-acetyltransferases, and methyltransferases, the activity and expression of which may be correlated with development. The expression of uridine diphosphate glucuronyltransferase in the liver is approximately 1% of adult levels during the GW of 30–40 and increases significantly to adult values by the first few weeks of life [84]. Different UGTs isoforms show different expression and activity patterns with increasing age, and all are found in the liver, early in gestation [12]. An LC-MS/MS proteomics method was used to investigate the ontogeny of six UGTs, and it was found that the protein abundances of UGT1A1, UGT1A4, UGT1A6, UGT1A9, UGT2B7, and UGT2B15 increased by approximately 8-, 55-, 35-, 33-, 8-, and 3-fold, respectively, from neonates to adults [85]. Similar to CYP450 enzymes, glucuronidation capacity maturation in neonates depends on PNA and postmenstrual age (PMA) rather than on GA [86]. Moreover, limited glucuronidated enzymes in neonates can be partly compensated by sulfate conjugation [87][88]. The expression of uridine 5′-diphospho-glucuronosyltransferase (SULTs) shows significantly different developmental patterns. For example, the protein expression of SULT1A1 did not change significantly during various developmental periods, whereas that of SULT2A1 increased during the third trimester of gestation and continued to increase after birth [89]. Information regarding the changes in the other phase II catalytic enzymes during development is presented in Table 3.
The drugs which are extracted greater than 70% by the liver are defined as high clearance drugs. Their intrinsic clearances are greater than liver blood flow. For these drugs, liver blood flow rather than enzymatic activity possesses a determinant effect on drug disposition [90]. Compared to adults, the fraction of hepatic blood flow in cardiac output is higher in children (38% vs. 24%) [91]. For neonates, hepatic function is slightly improved after birth because of an increase in neonatal hepatic blood flow related to pronounced alteration in postnatal circulatory systems [92].
Table 3. Ontogeny of phase I and phase II enzymes in human liver tissues. Fetus: 0 years, neonates: 0–1 months, infants: 1 month–2 years, children: 2–12 years, adolescents 12–16 years, adults: >16 years.

Abbreviations: ADH, alcohol or aldehyde dehydrogenases; AOX, aldehyde oxidase; CYP, cytochrome P450; EPHX, human cytosolic epoxide hydrolases; FMO, flavin-containing mono-oxygenase; GA, gestational age; GST, glutathione S-transferases; GW, gestational week; PNA, postnatal age; PON, paraoxonase; SULT, sulfotransferases; UGT, uridine 5′-diphospho-glucuronosyltransferase.

5. Drug Excretion

In addition to metabolic elimination, most drugs and their metabolites are eliminated from the body via kidneys. The kidneys in neonates are smaller than those in adults and continue to increase in size during the juvenile and pediatric periods [129]. This increase in size is mainly dependent on the increase in tubular mass because the number of glomeruli is constant from nephrogenesis to maturation [129]. The kidneys in preterm infants are much smaller than those in term newborns, and the mean total kidney volume doubles during PMA at 28–37 weeks [130]. Generally, human nephrogenesis and vasculogenesis occur in utero and are completed at GW 36, whereas tubule maturation and growth continue during the first year after birth [131][132]. Notably, nephrogenesis in preterm neonates lasts until 40 days after birth [31].
Renal clearance of drugs involves three processes: glomerular filtration, tubular secretion, and active/passive tubular reabsorption [133]. Renal elimination capacity depends on renal functional maturation, a dynamic process closely correlated with morphogenesis, with significant developmental changes occurring during gestation and the first 18 months of life. A delicate balance between the renal vasoconstrictive and vasodilatory forces leads to increased renal vascular resistance after birth [134]. Therefore, the renal blood flow in neonates is low, reaching only 10% of cardiac output by the first week of life [54]. Previous research has reported that adequate renal plasma flow in premature and term infants is 20 mL/min/1.73 m2 and 83 mL/min/1.73 m2, respectively [54]. Then, effective renal plasma flow increases with age, eventually reaching the adult levels of 650 mL/min/1.73 m2 by 2 years of age [54]. GFR changes dramatically following the decrease in renal vascular resistance and the increase in renal blood flow in early life [4][135]. It has been reported that GFR begins immediately after birth and is approximately 40% of adult values in neonates (41 ± 15 mL/min/1.73 m2) [4][135]. Subsequently, GRF increases rapidly to approximately 60% of adult values (66 ± 25 mL/min/1.73 m2) during PNA 2–8 weeks, reaching adult levels of 100–125 mL/min/1.73 m2 by the age of 8–12 months [4][135][136]. In addition, the GFR in preterm infants is only half of that of newborns compared to full-term infants, as nephrogenesis is incomplete until GW 34 and initially rises slowly to reach an average level until eight years of age [12][137]. Generally, the GFR is limited to newborns and appears to be dependent on weight, GA, and PNA [138]. The active tubular secretion is also immature in neonates and is approximately 20–30% of adult levels, and then approaches the adult capacity by 7–12 months of age [4][25]. Hence, the GFR matures more rapidly than tubular secretion, leading to glomerulo-tubular imbalance in neonates [138]. Tubular reabsorption is the last renal function to develop, displays the steepest rise during 1–3 years of age, and continually increases until adolescence [138][139]. Elimination through tubular secretion and reabsorption depends on renal blood flow [25]. Tubular secretion and reabsorption are involved in active transport processes, so maturational changes in transporters must be considered for drugs with extensive renal elimination.
Furthermore, biliary excretion is the main elimination pathway for some drugs, such as polar macromolecular compounds (>300 g/mol), oral drugs highly bound to hepatic transporters, and hydrophilic drugs that extensively bind to plasma proteins for parenteral administration [140]. Bile acid concentration in the intestine is low at birth, equivalent to approximately 60% of the adult levels, and increases to approximately 80% by one year of age [141]. The bile acid pool is reduced in both premature and full-term infants, and the full-term infants experience a remarkable expansion in the size of the bile acid pool at the end of pregnancy [142][143]. The bile acid pool then continues to increase with age, reaching adult levels by two years of age but approaching adult levels by seven weeks if corrected for body surface area [144]. In addition, numerous transporters are involved in drug transport across the basal outer membrane and tubule membrane of hepatocytes, along with bile acids; therefore, developmental changes in transporters are important for drug excretion through bile.

6. Transporters

Membrane transporters that are physiologically expressed throughout the body control the influx and efflux of endogenous and exogenous substances and affect the ADME process [145][146]. Investigating transporter ontogeny is challenging because of the lack of specific transporters and the unclear correlation between measured mRNA levels and actual protein expression. In general, the expression of transporters gradually increases during organogenesis [40][62][147][148]. The expression of P-glycoprotein, organic cation transporter 1 (OCT1), and organic anion-transporting polypeptide 1B3 is significantly lower at birth and increases with age. In contrast, the hepatic expression of multidrug resistance-associated protein 2 and OATP1B1 in neonates is delayed and reduced compared to that in adults until the first months of life [147][149][150]. In this research, researchers investigated the transporter expression in the liver, kidney, intestine, and blood–brain barrier during development (Table 4).
Table 4. The age-related changes in expression of membrane transporters in neonates. Fetus: 0 years, neonates: 0–1 months, infants: 1 month–2 years, children: 2–12 years, adolescents 12–16 years, adults: >16 years.

References

  1. Laughon, M.M.; Avant, D.; Tripathi, N.; Hornik, C.P.; Cohen-Wolkowiez, M.; Clark, R.H.; Smith, P.B.; Rodriguez, W. Drug labeling and exposure in neonates. JAMA Pediatr. 2014, 168, 130–136.
  2. Zhou, X.; Dun, J.; Chen, X.; Xiang, B.; Dang, Y.; Cao, D. Predicting the correct dose in children: Role of computational Pediatric Physiological-based pharmacokinetics modeling tools. CPT Pharmacomet. Syst. Pharmacol. 2023, 12, 13–26.
  3. Cella, M.; Knibbe, C.; Danhof, M.; Della Pasqua, O. What is the right dose for children? Br. J. Clin. Pharmacol. 2010, 70, 597–603.
  4. Kearns, G.L.; Abdel-Rahman, S.M.; Alander, S.W.; Blowey, D.L.; Leeder, J.S.; Kauffman, R.E. Developmental pharmacology--drug disposition, action, and therapy in infants and children. N. Engl. J. Med. 2003, 349, 1157–1167.
  5. Kaneria, N.S.; Tuleu, C.; Ernest, T. Opportunities for enteral drug delivery for neonates, infants, and toddlers: A critical exploration. Expert Opin. Drug Deliv. 2022, 19, 475–519.
  6. Smits, A.; Annaert, P.; Allegaert, K. Drug disposition and clinical practice in neonates: Cross talk between developmental physiology and pharmacology. Int. J. Pharm. 2013, 452, 8–13.
  7. Allegaert, K.; Abbasi, M.Y.; Michelet, R.; Olafuyi, O. The Impact of Low Cardiac Output on Propofol Pharmacokinetics across Age Groups-An Investigation Using Physiologically Based Pharmacokinetic Modelling. Pharmaceutics 2022, 14, 1957.
  8. Johnson, T.N.; Bonner, J.J.; Tucker, G.T.; Turner, D.B.; Jamei, M. Development and applications of a physiologically-based model of paediatric oral drug absorption. Eur. J. Pharm. Sci. 2018, 115, 57–67.
  9. Kohlmann, P.; Stillhart, C.; Kuentz, M.; Parrott, N. Investigating Oral Absorption of Carbamazepine in Pediatric Populations. AAPS J. 2017, 19, 1864–1877.
  10. Lu, H.; Rosenbaum, S. Developmental pharmacokinetics in pediatric populations. J. Pediatr. Pharmacol. Ther. 2014, 19, 262–276.
  11. Badee, J.; Qiu, N.; Parrott, N.; Collier, A.C.; Schmidt, S.; Fowler, S. Optimization of Experimental Conditions of Automated Glucuronidation Assays in Human Liver Microsomes Using a Cocktail Approach and Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry. Drug Metab. Dispos. 2019, 47, 124–134.
  12. Badee, J.; Fowler, S.; de Wildt, S.N.; Collier, A.C.; Schmidt, S.; Parrott, N. The Ontogeny of UDP-glucuronosyltransferase Enzymes, Recommendations for Future Profiling Studies and Application Through Physiologically Based Pharmacokinetic Modelling. Clin. Pharmacokinet. 2019, 58, 189–211.
  13. Wang, K.; Jiang, K.; Wei, X.; Li, Y.; Wang, T.; Song, Y. Physiologically Based Pharmacokinetic Models Are Effective Support for Pediatric Drug Development. AAPS PharmSciTech 2021, 22, 208.
  14. Van den Anker, J.; Reed, M.D.; Allegaert, K.; Kearns, G.L. Developmental Changes in Pharmacokinetics and Pharmacodynamics. J. Clin. Pharmacol. 2018, 58 (Suppl. 10), S10–S25.
  15. Mahmood, I. Prediction of Drug Clearance in Premature and Mature Neonates, Infants, and Children ≤ 2 Years of Age: A Comparison of the Predictive Performance of 4 Allometric Models. J. Clin. Pharmacol. 2016, 56, 733–739.
  16. Mahmood, I. Prediction of drug clearance in children: A review of different methodologies. Expert Opin. Drug Metab. Toxicol. 2015, 11, 573–587.
  17. Mahmood, I.; Staschen, C.M.; Goteti, K. Prediction of drug clearance in children: An evaluation of the predictive performance of several models. AAPS J. 2014, 16, 1334–1343.
  18. Mahmood, I. Dosing in children: A critical review of the pharmacokinetic allometric scaling and modelling approaches in paediatric drug development and clinical settings. Clin. Pharmacokinet. 2014, 53, 327–346.
  19. Edginton, A.N.; Shah, B.; Sevestre, M.; Momper, J.D. The integration of allometry and virtual populations to predict clearance and clearance variability in pediatric populations over the age of 6 years. Clin. Pharmacokinet. 2013, 52, 693–703.
  20. Mansoor, N.; Ahmad, T.; Alam Khan, R.; Sharib, S.M.; Mahmood, I. Prediction of Clearance and Dose of Midazolam in Preterm and Term Neonates: A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling. Am. J. Ther. 2019, 26, e32–e37.
  21. Johnson, T.N.; Rostami-Hodjegan, A.; Tucker, G.T. Prediction of the clearance of eleven drugs and associated variability in neonates, infants and children. Clin. Pharmacokinet. 2006, 45, 931–956.
  22. Leong, R.; Vieira, M.L.; Zhao, P.; Mulugeta, Y.; Lee, C.S.; Huang, S.M.; Burckart, G.J. Regulatory experience with physiologically based pharmacokinetic modeling for pediatric drug trials. Clin. Pharmacol. Ther. 2012, 91, 926–931.
  23. Johnson, T.N. The problems in scaling adult drug doses to children. Arch. Dis. Child. 2008, 93, 207–211.
  24. Carry, M.R.; Ringel, S.P.; Starcevich, J.M. Distribution of capillaries in normal and diseased human skeletal muscle. Muscle Nerve 1986, 9, 445–454.
  25. Tayman, C.; Rayyan, M.; Allegaert, K. Neonatal pharmacology: Extensive interindividual variability despite limited size. J. Pediatr. Pharmacol. Ther. 2011, 16, 170–184.
  26. Afsar, F.S. Physiological skin conditions of preterm and term neonates. Clin. Exp. Dermatol. 2010, 35, 346–350.
  27. Lin, Y.C.; Sussman, H.H.; Benitz, W.E. Plasma concentrations after rectal administration of acetaminophen in preterm neonates. Paediatr. Anaesth. 1997, 7, 457–459.
  28. Burri, P.H. Fetal and postnatal development of the lung. Annu. Rev. Physiol. 1984, 46, 617–628.
  29. El-Gendy, N.; Kaviratna, A.; Berkland, C.; Dhar, P. Delivery and performance of surfactant replacement therapies to treat pulmonary disorders. Ther. Deliv. 2013, 4, 951–980.
  30. Jamei, M.; Turner, D.; Yang, J.; Neuhoff, S.; Polak, S.; Rostami-Hodjegan, A.; Tucker, G. Population-based mechanistic prediction of oral drug absorption. AAPS J. 2009, 11, 225–237.
  31. Butranova, O.I.; Ushkalova, E.A.; Zyryanov, S.K.; Chenkurov, M.S. Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines 2023, 11, 940.
  32. Avery, G.B.; Randolph, J.G.; Weaver, T. Gastric acidity in the first day of life. Pediatrics 1966, 37, 1005–1007.
  33. Gan, J.; Bornhorst, G.M.; Henrick, B.M.; German, J.B. Protein Digestion of Baby Foods: Study Approaches and Implications for Infant Health. Mol. Nutr. Food Res. 2018, 62, 1700231.
  34. Indrio, F.; Neu, J.; Pettoello-Mantovani, M.; Marchese, F.; Martini, S.; Salatto, A.; Aceti, A. Development of the Gastrointestinal Tract in Newborns as a Challenge for an Appropriate Nutrition: A Narrative Review. Nutrients 2022, 14, 1405.
  35. Lebenthal, E.; Lee, P.C.; Heitlinger, L.A. Impact of development of the gastrointestinal tract on infant feeding. J. Pediatr. 1983, 102, 1–9.
  36. Verscheijden, L.F.M.; Koenderink, J.B.; Johnson, T.N.; de Wildt, S.N.; Russel, F.G.M. Physiologically-based pharmacokinetic models for children: Starting to reach maturation? Pharmacol. Ther. 2020, 211, 107541.
  37. Allegaert, K.; Mian, P.; van den Anker, J.N. Developmental Pharmacokinetics in Neonates: Maturational Changes and Beyond. Curr. Pharm. Des. 2017, 23, 5769–5778.
  38. Bourlieu, C.; Menard, O.; Bouzerzour, K.; Mandalari, G.; Macierzanka, A.; Mackie, A.R.; Dupont, D. Specificity of infant digestive conditions: Some clues for developing relevant in vitro models. Crit. Rev. Food Sci. Nutr. 2014, 54, 1427–1457.
  39. van Kalken, C.K.; Giaccone, G.; van der Valk, P.; Kuiper, C.M.; Hadisaputro, M.M.; Bosma, S.A.; Scheper, R.J.; Meijer, C.J.; Pinedo, H.M. Multidrug resistance gene (P-glycoprotein) expression in the human fetus. Am. J. Pathol. 1992, 141, 1063–1072.
  40. Mooij, M.G.; Schwarz, U.I.; de Koning, B.A.; Leeder, J.S.; Gaedigk, R.; Samsom, J.N.; Spaans, E.; van Goudoever, J.B.; Tibboel, D.; Kim, R.B.; et al. Ontogeny of human hepatic and intestinal transporter gene expression during childhood: Age matters. Drug Metab. Dispos. 2014, 42, 1268–1274.
  41. Linakis, M.W.; Roberts, J.K.; Lala, A.C.; Spigarelli, M.G.; Medlicott, N.J.; Reith, D.M.; Ward, R.M.; Sherwin, C.M. Challenges Associated with Route of Administration in Neonatal Drug Delivery. Clin. Pharmacokinet. 2016, 55, 185–196.
  42. Barbero, G.J.; Runge, G.; Fischer, D.; Crawford, M.N.; Torres, F.E.; Gyorgy, P. Investigations on the bacterial flora, pH, and sugar content in the intestinal tract of infants. J. Pediatr. 1952, 40, 152–163.
  43. Fredrikzon, B.; Olivecrona, T. Decrease of lipase and esterase activities in intestinal contents of newborn infants during test meals. Pediatr. Res. 1978, 12, 631–634.
  44. Nicolas, J.M.; Bouzom, F.; Hugues, C.; Ungell, A.L. Oral drug absorption in pediatrics: The intestinal wall, its developmental changes and current tools for predictions. Biopharm. Drug Dispos. 2017, 38, 209–230.
  45. van Elburg, R.M.; Fetter, W.P.; Bunkers, C.M.; Heymans, H.S. Intestinal permeability in relation to birth weight and gestational and postnatal age. Arch. Dis. Child. Fetal Neonatal Ed. 2003, 88, F52–F55.
  46. Scholtens, P.A.; Oozeer, R.; Martin, R.; Amor, K.B.; Knol, J. The early settlers: Intestinal microbiology in early life. Annu. Rev. Food Sci. Technol. 2012, 3, 425–447.
  47. de Waal, T.; Brouwers, J.; Mols, R.; Hoffman, I.; Rayyan, M.; Augustijns, P. Characterization of neonatal and infant enterostomy fluids. Int. J. Pharm. 2023, 639, 122943.
  48. Lebenthal, A.; Lebenthal, E. The ontogeny of the small intestinal epithelium. JPEN J. Parenter. Enteral Nutr. 1999, 23 (Suppl. 5), S3–S6.
  49. Lebenthal, E.; Lee, P.C. Development of functional responses in human exocrine pancreas. Pediatrics 1980, 66, 556–560.
  50. Carriere, F.; Barrowman, J.A.; Verger, R.; Laugier, R. Secretion and contribution to lipolysis of gastric and pancreatic lipases during a test meal in humans. Gastroenterology 1993, 105, 876–888.
  51. Lindower, J.B. Water balance in the fetus and neonate. Semin. Fetal Neonatal Med. 2017, 22, 71–75.
  52. Ward, R.M.; Benjamin, D.; Barrett, J.S.; Allegaert, K.; Portman, R.; Davis, J.M.; Turner, M.A. Safety, dosing, and pharmaceutical quality for studies that evaluate medicinal products (including biological products) in neonates. Pediatr. Res. 2017, 81, 692–711.
  53. Young, A.; Brown, L.K.; Ennis, S.; Beattie, R.M.; Johnson, M.J. Total body water in full-term and preterm newborns: Systematic review and meta-analysis. Arch. Dis. Child. Fetal Neonatal Ed. 2021, 106, 542–548.
  54. Allegaert, K.; Cossey, V.; van den Anker, J.N. Dosing Guidelines of Aminoglycosides in Neonates: A Balance Between Physiology and Feasibility. Curr. Pharm. Des. 2015, 21, 5699–5704.
  55. Allegaert, K.; van den Anker, J.N. Perinatal and neonatal use of paracetamol for pain relief. Semin. Fetal Neonatal Med. 2017, 22, 308–313.
  56. Schmidt, S.; Gonzalez, D.; Derendorf, H. Significance of protein binding in pharmacokinetics and pharmacodynamics. J. Pharm. Sci. 2010, 99, 1107–1122.
  57. Weaving, G.; Batstone, G.F.; Jones, R.G. Age and sex variation in serum albumin concentration: An observational study. Ann. Clin. Biochem. 2016, 53 Pt 1, 106–111.
  58. Chen, C.B.; Hammo, B.; Barry, J.; Radhakrishnan, K. Overview of Albumin Physiology and its Role in Pediatric Diseases. Curr. Gastroenterol. Rep. 2021, 23, 11.
  59. Anell-Olofsson, M.; Ahmadi, S.; Lonnqvist, P.A.; Eksborg, S.; von Horn, H.; Bartocci, M. Plasma concentrations of alpha-1-acid glycoprotein in preterm and term newborns: Influence of mode of delivery and implications for plasma protein binding of local anaesthetics. Br. J. Anaesth. 2018, 121, 427–431.
  60. Nau, H.; Luck, W.; Kuhnz, W. Decreased serum protein binding of diazepam and its major metabolite in the neonate during the first postnatal week relate to increased free fatty acid levels. Br. J. Clin. Pharmacol. 1984, 17, 92–98.
  61. Notarianni, L.J. Plasma protein binding of drugs in pregnancy and in neonates. Clin. Pharmacokinet. 1990, 18, 20–36.
  62. Yanni, S.B.; Smith, P.B.; Benjamin, D.K., Jr.; Augustijns, P.F.; Thakker, D.R.; Annaert, P.P. Higher clearance of micafungin in neonates compared with adults: Role of age-dependent micafungin serum binding. Biopharm. Drug Dispos. 2011, 32, 222–232.
  63. Pokorna, P.; Wildschut, E.D.; Vobruba, V.; van den Anker, J.N.; Tibboel, D. The Impact of Hypothermia on the Pharmacokinetics of Drugs Used in Neonates and Young Infants. Curr. Pharm. Des. 2015, 21, 5705–5724.
  64. Sethi, P.K.; White, C.A.; Cummings, B.S.; Hines, R.N.; Muralidhara, S.; Bruckner, J.V. Ontogeny of plasma proteins, albumin and binding of diazepam, cyclosporine, and deltamethrin. Pediatr. Res. 2016, 79, 409–415.
  65. McNamara, P.J.; Alcorn, J. Protein binding predictions in infants. AAPS PharmSci 2002, 4, 3.
  66. Alcorn, J.; McNamara, P.J. Ontogeny of hepatic and renal systemic clearance pathways in infants: Part I. Clin. Pharmacokinet. 2002, 41, 959–998.
  67. Sharer, J.E.; Wrighton, S.A. Identification of the human hepatic cytochromes P450 involved in the in vitro oxidation of antipyrine. Drug Metab. Dispos. 1996, 24, 487–494.
  68. Engel, G.; Hofmann, U.; Heidemann, H.; Cosme, J.; Eichelbaum, M. Antipyrine as a probe for human oxidative drug metabolism: Identification of the cytochrome P450 enzymes catalyzing 4-hydroxyantipyrine, 3-hydroxymethylantipyrine, and norantipyrine formation. Clin. Pharmacol. Ther. 1996, 59, 613–623.
  69. Allegaert, K.; Simons, S.H.P.; Tibboel, D.; Krekels, E.H.; Knibbe, C.A.; van den Anker, J.N. Non-maturational covariates for dynamic systems pharmacology models in neonates, infants, and children: Filling the gaps beyond developmental pharmacology. Eur. J. Pharm. Sci. 2017, 109S, S27–S31.
  70. Hines, R.N. Ontogeny of human hepatic cytochromes P450. J. Biochem. Mol. Toxicol. 2007, 21, 169–175.
  71. Mahmood, I.; Ahmad, T.; Mansoor, N.; Sharib, S.M. Prediction of Clearance in Neonates and Infants (≤3 Months of Age) for Drugs That Are Glucuronidated: A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling. J. Clin. Pharmacol. 2017, 57, 476–483.
  72. Johnson, T.N.; Rostami-Hodjegan, A. Resurgence in the use of physiologically based pharmacokinetic models in pediatric clinical pharmacology: Parallel shift in incorporating the knowledge of biological elements and increased applicability to drug development and clinical practice. Paediatr. Anaesth. 2011, 21, 291–301.
  73. Chen, H.; Brzezinski, M.R.; Fantel, A.G.; Juchau, M.R. Catalysis of drug oxidation during embryogenesis in human hepatic tissues using imipramine as a model substrate. Drug Metab. Dispos. 1999, 27, 1306–1308.
  74. Hakkola, J.; Raunio, H.; Purkunen, R.; Saarikoski, S.; Vahakangas, K.; Pelkonen, O.; Edwards, R.J.; Boobis, A.R.; Pasanen, M. Cytochrome P450 3A expression in the human fetal liver: Evidence that CYP3A5 is expressed in only a limited number of fetal livers. Biol. Neonate 2001, 80, 193–201.
  75. Lacroix, D.; Sonnier, M.; Moncion, A.; Cheron, G.; Cresteil, T. Expression of CYP3A in the human liver--evidence that the shift between CYP3A7 and CYP3A4 occurs immediately after birth. Eur. J. Biochem. 1997, 247, 625–634.
  76. de Wildt, S.N.; Kearns, G.L.; Leeder, J.S.; van den Anker, J.N. Cytochrome P450 3A: Ontogeny and drug disposition. Clin. Pharmacokinet. 1999, 37, 485–505.
  77. Ince, I.; Knibbe, C.A.; Danhof, M.; de Wildt, S.N. Developmental changes in the expression and function of cytochrome P450 3A isoforms: Evidence from in vitro and in vivo investigations. Clin. Pharmacokinet. 2013, 52, 333–345.
  78. Johnsrud, E.K.; Koukouritaki, S.B.; Divakaran, K.; Brunengraber, L.L.; Hines, R.N.; McCarver, D.G. Human hepatic CYP2E1 expression during development. J. Pharmacol. Exp. Ther. 2003, 307, 402–407.
  79. Maxwell, D.M. The specificity of carboxylesterase protection against the toxicity of organophosphorus compounds. Toxicol. Appl. Pharmacol. 1992, 114, 306–312.
  80. Pope, C.N.; Karanth, S.; Liu, J.; Yan, B. Comparative carboxylesterase activities in infant and adult liver and their in vitro sensitivity to chlorpyrifos oxon. Regul. Toxicol. Pharmacol. 2005, 42, 64–69.
  81. Yang, D.; Pearce, R.E.; Wang, X.; Gaedigk, R.; Wan, Y.J.; Yan, B. Human carboxylesterases HCE1 and HCE2: Ontogenic expression, inter-individual variability and differential hydrolysis of oseltamivir, aspirin, deltamethrin and permethrin. Biochem. Pharmacol. 2009, 77, 238–247.
  82. Zane, N.R.; Chen, Y.; Wang, M.Z.; Thakker, D.R. Cytochrome P450 and flavin-containing monooxygenase families: Age-dependent differences in expression and functional activity. Pediatr. Res. 2018, 83, 527–535.
  83. Tran, M.N.; Wu, A.H.; Hill, D.W. Alcohol dehydrogenase and catalase content in perinatal infant and adult livers: Potential influence on neonatal alcohol metabolism. Toxicol. Lett. 2007, 169, 245–252.
  84. Watchko, J.F.; Lin, Z. Exploring the genetic architecture of neonatal hyperbilirubinemia. Semin. Fetal Neonatal Med. 2010, 15, 169–175.
  85. Bhatt, D.K.; Mehrotra, A.; Gaedigk, A.; Chapa, R.; Basit, A.; Zhang, H.; Choudhari, P.; Boberg, M.; Pearce, R.E.; Gaedigk, R.; et al. Age- and Genotype-Dependent Variability in the Protein Abundance and Activity of Six Major Uridine Diphosphate-Glucuronosyltransferases in Human Liver. Clin. Pharmacol. Ther. 2019, 105, 131–141.
  86. Allegaert, K.; Vanhaesebrouck, S.; Verbesselt, R.; van den Anker, J.N. In vivo glucuronidation activity of drugs in neonates: Extensive interindividual variability despite their young age. Ther. Drug Monit. 2009, 31, 411–415.
  87. Levy, G.; Khanna, N.N.; Soda, D.M.; Tsuzuki, O.; Stern, L. Pharmacokinetics of acetaminophen in the human neonate: Formation of acetaminophen glucuronide and sulfate in relation to plasma bilirubin concentration and D-glucaric acid excretion. Pediatrics 1975, 55, 818–825.
  88. McRorie, T.I.; Lynn, A.M.; Nespeca, M.K.; Opheim, K.E.; Slattery, J.T. The maturation of morphine clearance and metabolism. Am. J. Dis. Child. 1992, 146, 972–976.
  89. Duanmu, Z.; Weckle, A.; Koukouritaki, S.B.; Hines, R.N.; Falany, J.L.; Falany, C.N.; Kocarek, T.A.; Runge-Morris, M. Developmental expression of aryl, estrogen, and hydroxysteroid sulfotransferases in pre- and postnatal human liver. J. Pharmacol. Exp. Ther. 2006, 316, 1310–1317.
  90. Nies, A.S.; Shand, D.G.; Wilkinson, G.R. Altered hepatic blood flow and drug disposition. Clin. Pharmacokinet. 1976, 1, 135–155.
  91. Bjorkman, S. Prediction of drug disposition in infants and children by means of physiologically based pharmacokinetic (PBPK) modelling: Theophylline and midazolam as model drugs. Br. J. Clin. Pharmacol. 2005, 59, 691–704.
  92. Gow, P.J.; Ghabrial, H.; Smallwood, R.A.; Morgan, D.J.; Ching, M.S. Neonatal hepatic drug elimination. Pharmacol. Toxicol. 2001, 88, 3–15.
  93. Shimada, T.; Yamazaki, H.; Mimura, M.; Wakamiya, N.; Ueng, Y.F.; Guengerich, F.P.; Inui, Y. Characterization of microsomal cytochrome P450 enzymes involved in the oxidation of xenobiotic chemicals in human fetal liver and adult lungs. Drug Metab. Dispos. 1996, 24, 515–522.
  94. Yang, H.Y.; Namkung, M.J.; Juchau, M.R. Expression of functional cytochrome P4501A1 in human embryonic hepatic tissues during organogenesis. Biochem. Pharmacol. 1995, 49, 717–726.
  95. Omiecinski, C.J.; Redlich, C.A.; Costa, P. Induction and developmental expression of cytochrome P450IA1 messenger RNA in rat and human tissues: Detection by the polymerase chain reaction. Cancer Res. 1990, 50, 4315–4321.
  96. Sonnier, M.; Cresteil, T. Delayed ontogenesis of CYP1A2 in the human liver. Eur. J. Biochem. 1998, 251, 893–898.
  97. Shimada, T.; Yamazaki, H.; Mimura, M.; Inui, Y.; Guengerich, F.P. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: Studies with liver microsomes of 30 Japanese and 30 Caucasians. J. Pharmacol. Exp. Ther. 1994, 270, 414–423.
  98. Hakkola, J.; Pasanen, M.; Pelkonen, O.; Hukkanen, J.; Evisalmi, S.; Anttila, S.; Rane, A.; Mantyla, M.; Purkunen, R.; Saarikoski, S.; et al. Expression of CYP1B1 in human adult and fetal tissues and differential inducibility of CYP1B1 and CYP1A1 by Ah receptor ligands in human placenta and cultured cells. Carcinogenesis 1997, 18, 391–397.
  99. Tateishi, T.; Nakura, H.; Asoh, M.; Watanabe, M.; Tanaka, M.; Kumai, T.; Takashima, S.; Imaoka, S.; Funae, Y.; Yabusaki, Y.; et al. A comparison of hepatic cytochrome P450 protein expression between infancy and postinfancy. Life Sci. 1997, 61, 2567–2574.
  100. Hakkola, J.; Pasanen, M.; Purkunen, R.; Saarikoski, S.; Pelkonen, O.; Maenpaa, J.; Rane, A.; Raunio, H. Expression of xenobiotic-metabolizing cytochrome P450 forms in human adult and fetal liver. Biochem. Pharmacol. 1994, 48, 59–64.
  101. Treluyer, J.M.; Gueret, G.; Cheron, G.; Sonnier, M.; Cresteil, T. Developmental expression of CYP2C and CYP2C-dependent activities in the human liver: In-vivo/in-vitro correlation and inducibility. Pharmacogenetics 1997, 7, 441–452.
  102. Yanni, S.B.; Annaert, P.P.; Augustijns, P.; Ibrahim, J.G.; Benjamin, D.K., Jr.; Thakker, D.R. In vitro hepatic metabolism explains higher clearance of voriconazole in children versus adults: Role of CYP2C19 and flavin-containing monooxygenase 3. Drug Metab. Dispos. 2010, 38, 25–31.
  103. Treluyer, J.M.; Jacqz-Aigrain, E.; Alvarez, F.; Cresteil, T. Expression of CYP2D6 in developing human liver. Eur. J. Biochem. 1991, 202, 583–588.
  104. Vieira, I.; Sonnier, M.; Cresteil, T. Developmental expression of CYP2E1 in the human liver. Hypermethylation control of gene expression during the neonatal period. Eur. J. Biochem. 1996, 238, 476–483.
  105. Carpenter, S.P.; Lasker, J.M.; Raucy, J.L. Expression, induction, and catalytic activity of the ethanol-inducible cytochrome P450 (CYP2E1) in human fetal liver and hepatocytes. Mol. Pharmacol. 1996, 49, 260–268.
  106. Boutelet-Bochan, H.; Huang, Y.; Juchau, M.R. Expression of CYP2E1 during embryogenesis and fetogenesis in human cephalic tissues: Implications for the fetal alcohol syndrome. Biochem. Biophys. Res. Commun. 1997, 238, 443–447.
  107. Gu, J.; Su, T.; Chen, Y.; Zhang, Q.Y.; Ding, X. Expression of biotransformation enzymes in human fetal olfactory mucosa: Potential roles in developmental toxicity. Toxicol. Appl. Pharmacol. 2000, 165, 158–162.
  108. Chen, Y.T.; Trzoss, L.; Yang, D.; Yan, B. Ontogenic expression of human carboxylesterase-2 and cytochrome P450 3A4 in liver and duodenum: Postnatal surge and organ-dependent regulation. Toxicology 2015, 330, 55–61.
  109. Schuetz, J.D.; Beach, D.L.; Guzelian, P.S. Selective expression of cytochrome P450 CYP3A mRNAs in embryonic and adult human liver. Pharmacogenetics 1994, 4, 11–20.
  110. Cresteil, T. Onset of xenobiotic metabolism in children: Toxicological implications. Food Addit. Contam. 1998, 15, 45–51.
  111. Koukouritaki, S.B.; Simpson, P.; Yeung, C.K.; Rettie, A.E.; Hines, R.N. Human hepatic flavin-containing monooxygenases 1 (FMO1) and 3 (FMO3) developmental expression. Pediatr. Res. 2002, 51, 236–243.
  112. Smith, M.; Hopkinson, D.A.; Harris, H. Developmental changes and polymorphism in human alcohol dehydrogenase. Ann. Hum. Genet. 1971, 34, 251–271.
  113. Estonius, M.; Svensson, S.; Hoog, J.O. Alcohol dehydrogenase in human tissues: Localisation of transcripts coding for five classes of the enzyme. FEBS Lett. 1996, 397, 338–342.
  114. Omiecinski, C.J.; Aicher, L.; Swenson, L. Developmental expression of human microsomal epoxide hydrolase. J. Pharmacol. Exp. Ther. 1994, 269, 417–423.
  115. Cresteil, T.; Beaune, P.; Kremers, P.; Celier, C.; Guengerich, F.P.; Leroux, J.P. Immunoquantification of epoxide hydrolase and cytochrome P-450 isozymes in fetal and adult human liver microsomes. Eur. J. Biochem. 1985, 151, 345–350.
  116. Mackness, B.; Beltran-Debon, R.; Aragones, G.; Joven, J.; Camps, J.; Mackness, M. Human tissue distribution of paraoxonases 1 and 2 mRNA. IUBMB Life 2010, 62, 480–482.
  117. Tayama, Y.; Sugihara, K.; Sanoh, S.; Miyake, K.; Kitamura, S.; Ohta, S. Developmental changes of aldehyde oxidase activity and protein expression in human liver cytosol. Drug Metab. Pharmacokinet. 2012, 27, 543–547.
  118. Strassburg, C.P.; Strassburg, A.; Kneip, S.; Barut, A.; Tukey, R.H.; Rodeck, B.; Manns, M.P. Developmental aspects of human hepatic drug glucuronidation in young children and adults. Gut 2002, 50, 259–265.
  119. Zaya, M.J.; Hines, R.N.; Stevens, J.C. Epirubicin glucuronidation and UGT2B7 developmental expression. Drug Metab. Dispos. 2006, 34, 2097–2101.
  120. Bhatt, D.K.; Basit, A.; Zhang, H.; Gaedigk, A.; Lee, S.B.; Claw, K.G.; Mehrotra, A.; Chaudhry, A.S.; Pearce, R.E.; Gaedigk, R.; et al. Hepatic Abundance and Activity of Androgen- and Drug-Metabolizing Enzyme UGT2B17 Are Associated with Genotype, Age, and Sex. Drug Metab. Dispos. 2018, 46, 888–896.
  121. Richard, K.; Hume, R.; Kaptein, E.; Stanley, E.L.; Visser, T.J.; Coughtrie, M.W. Sulfation of thyroid hormone and dopamine during human development: Ontogeny of phenol sulfotransferases and arylsulfatase in liver, lung, and brain. J. Clin. Endocrinol. Metab. 2001, 86, 2734–2742.
  122. Stanley, E.L.; Hume, R.; Coughtrie, M.W. Expression profiling of human fetal cytosolic sulfotransferases involved in steroid and thyroid hormone metabolism and in detoxification. Mol. Cell. Endocrinol. 2005, 240, 32–42.
  123. Her, C.; Kaur, G.P.; Athwal, R.S.; Weinshilboum, R.M. Human sulfotransferase SULT1C1: cDNA cloning, tissue-specific expression, and chromosomal localization. Genomics 1997, 41, 467–470.
  124. Strange, R.C.; Davis, B.A.; Faulder, C.G.; Cotton, W.; Bain, A.D.; Hopkinson, D.A.; Hume, R. The human glutathione S-transferases: Developmental aspects of the GST1, GST2, and GST3 loci. Biochem. Genet. 1985, 23, 1011–1028.
  125. Raijmakers, M.T.; Steegers, E.A.; Peters, W.H. Glutathione S-transferases and thiol concentrations in embryonic and early fetal tissues. Hum. Reprod. 2001, 16, 2445–2450.
  126. Strange, R.C.; Howie, A.F.; Hume, R.; Matharoo, B.; Bell, J.; Hiley, C.; Jones, P.; Beckett, G.J. The development expression of alpha-, mu- and pi-class glutathione S-transferases in human liver. Biochim. Biophys. Acta 1989, 993, 186–190.
  127. Beckett, G.J.; Howie, A.F.; Hume, R.; Matharoo, B.; Hiley, C.; Jones, P.; Strange, R.C. Human glutathione S-transferases: Radioimmunoassay studies on the expression of alpha-, mu- and pi-class isoenzymes in developing lung and kidney. Biochim. Biophys. Acta 1990, 1036, 176–182.
  128. Li, W.; Gu, Y.; James, M.O.; Hines, R.N.; Simpson, P.; Langaee, T.; Stacpoole, P.W. Prenatal and postnatal expression of glutathione transferase zeta 1 in human liver and the roles of haplotype and subject age in determining activity with dichloroacetate. Drug Metab. Dispos. 2012, 40, 232–239.
  129. Frazier, K.S. Species Differences in Renal Development and Associated Developmental Nephrotoxicity. Birth Defects Res. 2017, 109, 1243–1256.
  130. Kandasamy, Y.; Rudd, D.; Smith, R.; Lumbers, E.R.; Wright, I.M. Extra uterine development of preterm kidneys. Pediatr. Nephrol. 2018, 33, 1007–1012.
  131. Solhaug, M.J.; Bolger, P.M.; Jose, P.A. The developing kidney and environmental toxins. Pediatrics 2004, 113 (Suppl. 4), 1084–1091.
  132. Bueters, R.; Bael, A.; Gasthuys, E.; Chen, C.; Schreuder, M.F.; Frazier, K.S. Ontogeny and Cross-species Comparison of Pathways Involved in Drug Absorption, Distribution, Metabolism, and Excretion in Neonates (Review): Kidney. Drug Metab. Dispos. 2020, 48, 353–367.
  133. Mahmood, I. Prediction of total and renal clearance of renally secreted drugs in neonates and infants (≤3 months of age). J. Clin. Transl. Res. 2022, 8, 445–452.
  134. Filler, G.; Bhayana, V.; Schott, C.; Diaz-Gonzalez de Ferris, M.E. How should we assess renal function in neonates and infants? Acta Paediatr. 2021, 110, 773–780.
  135. Loebstein, R.; Koren, G. Clinical pharmacology and therapeutic drug monitoring in neonates and children. Pediatr. Rev. 1998, 19, 423–428.
  136. Rhodin, M.M.; Anderson, B.J.; Peters, A.M.; Coulthard, M.G.; Wilkins, B.; Cole, M.; Chatelut, E.; Grubb, A.; Veal, G.J.; Keir, M.J.; et al. Human renal function maturation: A quantitative description using weight and postmenstrual age. Pediatr. Nephrol. 2009, 24, 67–76.
  137. Iacobelli, S.; Loprieno, S.; Bonsante, F.; Latorre, G.; Esposito, L.; Gouyon, J.B. Renal function in early childhood in very low birthweight infants. Am. J. Perinatol. 2007, 24, 587–592.
  138. Zhang, Y.; Mehta, N.; Muhari-Stark, E.; Burckart, G.J.; van den Anker, J.; Wang, J. Pediatric Renal Ontogeny and Applications in Drug Development. J. Clin. Pharmacol. 2019, 59 (Suppl. 1), S9–S20.
  139. van den Anker, J.; Allegaert, K. Considerations for Drug Dosing in Premature Infants. J. Clin. Pharmacol. 2021, 61 (Suppl. 1), S141–S151.
  140. Thabit, A.K. Antibiotics in the Biliary Tract: A Review of the Pharmacokinetics and Clinical Outcomes of Antibiotics Penetrating the Bile and Gallbladder Wall. Pharmacotherapy 2020, 40, 672–691.
  141. Balistreri, W.F. Immaturity of hepatic excretory function and the ontogeny of bile acid metabolism. J. Pediatr. Gastroenterol. Nutr. 1983, 2 (Suppl. 1), S207–S214.
  142. Subbiah, M.T.; Hassan, A.S. Development of bile acid biogenesis and its significance in cholesterol homeostasis. Adv. Lipid Res. 1982, 19, 137–161.
  143. Grijalva, J.; Vakili, K. Neonatal liver physiology. Semin. Pediatr. Surg. 2013, 22, 185–189.
  144. Heubi, J.E.; Balistreri, W.F.; Suchy, F.J. Bile salt metabolism in the first year of life. J. Lab. Clin. Med. 1982, 100, 127–136.
  145. Ho, R.H.; Kim, R.B. Transporters and drug therapy: Implications for drug disposition and disease. Clin. Pharmacol. Ther. 2005, 78, 260–277.
  146. Shitara, Y.; Sato, H.; Sugiyama, Y. Evaluation of drug-drug interaction in the hepatobiliary and renal transport of drugs. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 689–723.
  147. Konieczna, A.; Erdosova, B.; Lichnovska, R.; Jandl, M.; Cizkova, K.; Ehrmann, J. Differential expression of ABC transporters (MDR1, MRP1, BCRP) in developing human embryos. J. Mol. Histol. 2011, 42, 567–574.
  148. Kumar, V.; Prasad, B.; Patilea, G.; Gupta, A.; Salphati, L.; Evers, R.; Hop, C.E.; Unadkat, J.D. Quantitative transporter proteomics by liquid chromatography with tandem mass spectrometry: Addressing methodologic issues of plasma membrane isolation and expression-activity relationship. Drug Metab. Dispos. 2015, 43, 284–288.
  149. Klaassen, C.D.; Aleksunes, L.M. Xenobiotic, bile acid, and cholesterol transporters: Function and regulation. Pharmacol. Rev. 2010, 62, 1–96.
  150. Deo, A.K.; Prasad, B.; Balogh, L.; Lai, Y.; Unadkat, J.D. Interindividual variability in hepatic expression of the multidrug resistance-associated protein 2 (MRP2/ABCC2): Quantification by liquid chromatography/tandem mass spectrometry. Drug Metab. Dispos. 2012, 40, 852–855.
  151. Prasad, B.; Gaedigk, A.; Vrana, M.; Gaedigk, R.; Leeder, J.S.; Salphati, L.; Chu, X.; Xiao, G.; Hop, C.; Evers, R.; et al. Ontogeny of Hepatic Drug Transporters as Quantified by LC-MS/MS Proteomics. Clin. Pharmacol. Ther. 2016, 100, 362–370.
  152. van Groen, B.D.; van de Steeg, E.; Mooij, M.G.; van Lipzig, M.M.H.; de Koning, B.A.E.; Verdijk, R.M.; Wortelboer, H.M.; Gaedigk, R.; Bi, C.; Leeder, J.S.; et al. Proteomics of human liver membrane transporters: A focus on fetuses and newborn infants. Eur. J. Pharm. Sci. 2018, 124, 217–227.
  153. Abanda, N.N.; Riches, Z.; Collier, A.C. Lobular Distribution and Variability in Hepatic ATP Binding Cassette Protein B1 (ABCB1, P-gp): Ontogenetic Differences and Potential for Toxicity. Pharmaceutics 2017, 9, 8.
  154. Mooij, M.G.; van de Steeg, E.; van Rosmalen, J.; Windster, J.D.; de Koning, B.A.; Vaes, W.H.; van Groen, B.D.; Tibboel, D.; Wortelboer, H.M.; de Wildt, S.N. Proteomic Analysis of the Developmental Trajectory of Human Hepatic Membrane Transporter Proteins in the First Three Months of Life. Drug Metab. Dispos. 2016, 44, 1005–1013.
  155. Burgess, K.S.; Philips, S.; Benson, E.A.; Desta, Z.; Gaedigk, A.; Gaedigk, R.; Segar, M.W.; Liu, Y.; Skaar, T.C. Age-Related Changes in MicroRNA Expression and Pharmacogenes in Human Liver. Clin. Pharmacol. Ther. 2015, 98, 205–215.
  156. Sharma, S.; Ellis, E.C.; Gramignoli, R.; Dorko, K.; Tahan, V.; Hansel, M.; Mattison, D.R.; Caritis, S.N.; Hines, R.N.; Venkataramanan, R.; et al. Hepatobiliary disposition of 17-OHPC and taurocholate in fetal human hepatocytes: A comparison with adult human hepatocytes. Drug Metab. Dispos. 2013, 41, 296–304.
  157. Hahn, D.; Emoto, C.; Vinks, A.A.; Fukuda, T. Developmental Changes in Hepatic Organic Cation Transporter OCT1 Protein Expression from Neonates to Children. Drug Metab. Dispos. 2017, 45, 23–26.
  158. Chen, H.L.; Chen, H.L.; Liu, Y.J.; Feng, C.H.; Wu, C.Y.; Shyu, M.K.; Yuan, R.H.; Chang, M.H. Developmental expression of canalicular transporter genes in human liver. J. Hepatol. 2005, 43, 472–477.
  159. Cheung, K.W.K.; van Groen, B.D.; Spaans, E.; van Borselen, M.D.; de Bruijn, A.; Simons-Oosterhuis, Y.; Tibboel, D.; Samsom, J.N.; Verdijk, R.M.; Smeets, B.; et al. A Comprehensive Analysis of Ontogeny of Renal Drug Transporters: mRNA Analyses, Quantitative Proteomics, and Localization. Clin. Pharmacol. Ther. 2019, 106, 1083–1092.
  160. Lam, J.; Baello, S.; Iqbal, M.; Kelly, L.E.; Shannon, P.T.; Chitayat, D.; Matthews, S.G.; Koren, G. The ontogeny of P-glycoprotein in the developing human blood-brain barrier: Implication for opioid toxicity in neonates. Pediatr. Res. 2015, 78, 417–421.
  161. Miki, Y.; Suzuki, T.; Tazawa, C.; Blumberg, B.; Sasano, H. Steroid and xenobiotic receptor (SXR), cytochrome P450 3A4 and multidrug resistance gene 1 in human adult and fetal tissues. Mol. Cell. Endocrinol. 2005, 231, 75–85.
More
Information
Subjects: Pediatrics
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 117
Revisions: 2 times (View History)
Update Date: 25 Dec 2023
1000/1000